You are on page 1of 28

FEMS Microbiology Reviews, fuz016, 43, 2019, 548–575

doi: 10.1093/femsre/fuz016
Advance Access Publication Date: 10 June 2019
Review article

REVIEW ARTICLE

Cell wall peptidoglycan in Mycobacterium tuberculosis:


An Achilles’ heel for the TB-causing pathogen
Arundhati Maitra1,† , Tulika Munshi1,‡,† , Jess Healy2 , Liam T. Martin1 ,
Waldemar Vollmer3 , Nicholas H. Keep1 and Sanjib Bhakta1, *
1
Mycobacteria Research Laboratory, Institute of Structural and Molecular Biology, Department of Biological
Sciences, Birkbeck, University of London, Malet Street, London WC1E 7HX, UK, 2 Department of
Pharmaceutical and Biological Chemistry, UCL School of Pharmacy, 29–39 Brunswick Square, London WC1N
1AX, UK and 3 The Centre for Bacterial Cell Biology, Institute for Cell and Molecular Biosciences, Newcastle
University, Richardson Road, Newcastle upon Tyne, NE2 4AX, UK

Corresponding author: Mycobacteria Research Laboratory, Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck,
University of London, Malet Street, London WC1E 7HX, UK. Tel: +44-0207631-6355; Fax: +44-0207631-6246; E-mail: sanjib.bhakta@ucl.ac.uk
One sentence summary: A comprehensive, investigative review that highlights key enzymes involved in mycobacterial cell wall peptidoglycan
metabolism as novel anti-infective drug targets.

Equal contribution

Current address: Infection & Immunity, St. George’s, University of London, Cranmer Terrace, London SW17 0RE, UK
Editor: Chris Whitfield

ABSTRACT
Tuberculosis (TB), caused by the intracellular pathogen Mycobacterium tuberculosis, remains one of the leading causes of
mortality across the world. There is an urgent requirement to build a robust arsenal of effective antimicrobials, targeting
novel molecular mechanisms to overcome the challenges posed by the increase of antibiotic resistance in TB. Mycobacterium
tuberculosis has a unique cell envelope structure and composition, containing a peptidoglycan layer that is essential for
maintaining cellular integrity and for virulence. The enzymes involved in the biosynthesis, degradation, remodelling and
recycling of peptidoglycan have resurfaced as attractive targets for anti-infective drug discovery. Here, we review the
importance of peptidoglycan, including the structure, function and regulation of key enzymes involved in its metabolism.
We also discuss known inhibitors of ATP-dependent Mur ligases, and discuss the potential for the development of
pan-enzyme inhibitors targeting multiple Mur ligases.

Keywords: Tuberculosis (TB); cell envelope; peptidoglycan; metabolism; drug-target validation; antibiotic resistance;
antibacterial

INTRODUCTION million deaths reported due to TB in 2017 alone (WHO 2018).


An estimated 457 000 cases reported in 2017 presently harbour
Tuberculosis (TB) is a leading cause of mortality in the world
multi-drug resistant TB (MDR-TB), 8.5% of which are expected to
today and is caused by the bacterial pathogen Mycobacterium
be extensively drug resistant TB (XDR-TB) (WHO 2018), reflect-
tuberculosis. Despite innovations in diagnostics and improved
ing the urgent need to design and develop novel drugs to
access to health care, the global burden of TB remains sub-
treat TB.
stantial with around 10 million new cases of infection and 1.6

Received: 7 March 2019; Accepted: 7 June 2019


C The Author(s) 2019. Published by Oxford University Press on behalf of FEMS. This is an Open Access article distributed under the terms of the

Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and
reproduction in any medium, provided the original work is properly cited.

548
Maitra et al. 549

The success of M. tuberculosis as a pathogen and its innate acid (MurNAc) residues. Adjacent glycan strands are con-
resistance to many antimicrobial drugs can be attributed in part nected by short peptide stems with the sequence l-alanyl-
to its unique cell wall structure, which has low permeability for γ -d-isoglutamyl-meso-diaminopimelate-d-alanyl-d-alanine and
many drugs and possesses a large number of efflux pumps (Jar- linked to the d-lactoyl residue of each MurNAc. Peptides protrud-
lier and Nikaido 1994, Brennan and Nikaido 1995). The cell wall ing from adjacent glycan strands can be cross-linked between
is a defining characteristic of all bacteria. Amongst the many the carboxyl group of d-Ala at the fourth position and the ε-
purposes it serves, maintaining the cell-shape and withstanding amino group of the meso-diaminopimelate (mDAP) in the third
turgor are key. The varying properties of the bacterial cell wall, position.
especially the thickness of the peptidoglycan (PG), impart differ- While the basic structure of PG is conserved, it is important
ent stain-retention properties to the bacterial cell and enable us to note that the PG in mycobacteria undergoes several modifi-
to categorise most bacteria into Gram-positive, Gram-negative cations (Fig. 1B). PG isolated from M. tuberculosis and M. smeg-
and acid-fast. The presence of PG across nearly all bacteria indi- matis contains both N-acetylmuramic acid (MurNAc) and N-
cates that it was likely to have been present in their last common glycolylmuramic acid (MurNGlyc), whereas most other bacte-
ancestor (Errington 2013). Importantly, PG is essential for bacte- ria, including M. leprae, exclusively contain MurNAc (Mahap-
rial cell survival in most environments, thus making it a good atra et al. 2005a, Petit et al. 1969; Mahapatra et al. 2008). The
target for anti-infective therapy. presence of MurNGlyc increases the resistance of mycobacterial
Mycobacteria belong to the diverse family of Actinobac- PG to lysozyme (Raymond et al. 2005). Modifications such as N-
teria. The main components of the mycobacterial cell wall deacetylation or O-acetylation of GlcNAc and MurNAc respec-
are the PG layer, mycolic acid (MA) and arabinogalactan (AG). tively, as observed in other pathogenic bacteria (Vollmer 2008)
The mycobacterial cell wall resembles both the Gram-positive have not been reported in mycobacteria. Consequently, the dele-
and Gram-negative cell envelope by having a PG layer nearly tion of the hydroxylase responsible for N-glycolylation, namH,
as thick as the former and an outer, waxy layer mimicking increases the sensitivity towards β-lactams and lysozyme in M.
the outer membrane of the latter (Fig. 1A). The cell wall smegmatis (Raymond et al. 2005). Glycolylated PG fragments are
of mycobacteria plays a key role in intrinsic antibiotic resis- more potent at inducing NOD2-mediated host responses such
tance and virulence (Forrellad et al. 2013; Becker and Sander as production of tumour necrosis factor α (Hansen et al. 2014),
2016) but it is unclear why and how its complex structure which may appear to be counter-productive for the success of
evolved. Vincent et al. recently proposed that the ‘mycomem- an intracellular pathogen. However, the enhanced stimulation of
brane’ evolved by successive horizontal acquisition of genes; the host might enable M. tuberculosis to recruit phagocytic cells,
explaining the narrow distribution of the AG and MA biosyn- its ecological niche within the host and facilitate transmission
thetic genes in some lineages of the Actinobacteria that evolved to a new host.
later, while the PG genes are broadly distributed (Vincent et al. The peptide stems in PG undergo modifications such as ami-
2018). dation of the α-carboxylic group of d-isoglutamate (d-iGlu) and
PG is a dynamic structure but the factors that regulate its the ε-carboxylic group of the mDAP residues (Kotani et al. 1970;
composition and biogenesis in the slow-growing intra-cellular Mahapatra et al. 2008). d-iGlu amidation is required for the
pathogen M. tuberculosis are not well understood. The mycobac- functioning of PG transpeptidases that cross-link the peptides
terial PG plays a key role in the cell’s growth, cell–cell communi- to form a robust PG (Figueiredo et al. 2012; Zapun et al. 2013)
cation and in the initiation of the host immune response. The whereas mDAP amidation is involved in modulating PG hydrol-
cell envelope of some model bacteria such as Escherichia coli, ysis thereby maintaining the integrity of the cell wall (Nakel,
have long been the focus of extensive research; however, these Ghuysen and Kandler 1971; Dajkovic et al. 2017). These modi-
organisms do not serve as appropriate models when studying fications reduce the net negative charge of the cell wall, which
the unique physiology and biochemistry of M. tuberculosis. Here, impairs the efficacy of antimicrobial factors such as lysozyme,
we review the different stages of PG metabolism in M. tubercu- antimicrobial peptides and cell wall acting antibiotics (Girardin
losis, its importance for survival of the bacilli and the potential et al. 2003b; Roychowdhury, Wolfert and Boons 2005). In the PG
therapeutic targets it offers for the design of new anti-infective of M. leprae, Gly replaces l-Ala in position 1 of the peptide stem,
drugs. and an additional Gly residue is attached to mDAP (Mahapatra
et al. 2008).
The PG of M. tuberculosis contains a particularly high percent-
age (70%–80%) of cross-linked peptides (Wietzerbin et al. 1974),
compared to 40%–50% in E. coli (Glauner, Holtje and Schwarz
Modifications of cell wall PG in M. tuberculosis
1988). One-third of the peptide cross-links are DD-(or 4→3)
Lying outside the cytoplasmic membrane of M. tuberculosis, the bonds between d-Ala and mDAP, and about two-thirds are LD-(or
PG layer is covalently attached to AG which itself serves as 3→3) bonds linking two mDAP residues (Wietzerbin et al. 1974;
an attachment site for unique MAs (Fig. 1A). This is referred Hett and Rubin 2008) (Fig. 1B). Although LD-crosslinks are rela-
to as the cell wall core, or as the MA-AG-PG complex (MAPc) tively rare in many other bacterial species, recent reports have
(Brennan 2003). Beyond and interspersed within the MAPc estimated them to account for 80% of the peptide cross-links
are free lipids, phosphatidyl inositol mannosides (PIM), lipo- in the PG of stationary phase or dormant cells of M. tuberculo-
mannans (LM) and lipoarabinomannans (LAM). As observed sis (Lavollay et al. 2008). The LD-cross-links are also enhanced to
in other microorganisms (Jankute et al. 2015), the disruption 15%–20% of all cross-links in the PG of stationary phase cells of E.
of PG synthesis or digestion of the PG layer leads to the for- coli, but are significantly reduced (5% of all cross-links) in expo-
mation of L-form bacilli that display a variety of cell shapes nentially growing cells (Pisabarro, de Pedro and Vazquez 1985;
and morphology in mycobacteria (Udou, Ogawa and Mizuguchi Glauner, Holtje and Schwarz 1988; Templin, Ursinus and Holtje
1982). 1999).
PG is made of glycan strands of alternating, β(1→4)- Interestingly, the pattern of PG incorporation into the cell
linked N-acetylglucosamine (GlcNAc) and N-acetylmuramic wall of M. tuberculosis is different compared to the PG of other
550 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

Figure 1. (A), Differences in the cell envelope architecture of Gram-positive, acid-fast and Gram-negative bacteria. AG, arabinogalactan; GL, glycolipid; LAM, lipoarabino-
mannan; LP, lipoprotein; LPS, lipopolysaccharide; LTA, lipoteichoic acid; MA, mycolic acid; MAP, membrane-associated protein; OM, outer membrane; PG, peptidoglycan;
PM, plasma membrane; TA, teichoic acid . (B), PG structure revealing the unique features present in mycobacterial cells highlighted in blue. The stars depict residues
that undergo amidation.

rod-shaped bacteria (Daniel and Errington 2003; Hett and Rubin Imaging PG
2008). Most rod-shaped bacilli such as E. coli and Bacillus subtilis
elongate by inserting nascent PG along the lateral sides of the The recent application of fluorescence and atomic force
cell (den Blaauwen et al. 2008). An actin-like protein, MreB, posi- microscopy techniques together with new probes for labelling
tions the PG-elongation machinery along the short axis of the have significantly advanced our understanding of the dynamics
cell so that glycan strands are inserted circumferentially thus of PG synthesis (Radkov et al. 2018). Localisation studies using
maintaining the rod-shape (Domı́nguez-Escobar et al. 2011). In GFP-labelled PG biosynthetic proteins (Joyce et al. 2012; Grze-
contrast, mycobacteria lack MreB and other proteins important gorzewicz et al. 2016), fluorescent d-amino acids (Siegrist et al.
for lateral wall elongation and instead incorporate nascent PG at 2013; Meniche et al. 2014; Boutte et al. 2016; Schubert et al. 2017;
both poles of the cell (Thanky, Young and Robertson 2007; Kieser Botella et al. 2017b; Rodriguez-Rivera et al. 2018) and cell wall
and Rubin 2014). Mycobacteria exhibit differential incorporation targeting antibiotics (Thanky, Young and Robertson 2007; Kas-
of PG at the two cell poles (old versus new cell pole) at certain trinsky and Barry 2010) have further emphasized the impor-
stages of the cell cycle (before/after cytokinesis); this allows for tance of PG remodelling in cell elongation, septum formation
asymmetric cell division and results in a heterogeneous popu- and cell division. Aldridge et al. used microfluidics-based live-
lation of cells (Kieser and Rubin 2014; Botella et al. 2017b). Bara- cell imaging technique to show that the heterogeneous daugh-
nowski et al. reported that the ability of the mycobacterial cell to ter cell populations differ in their susceptibility to antibiotics
generate LD-cross-links (discussed later in this review) is essen- (Aldridge et al. 2012). Botella et al. compared the spatiotempo-
tial in maintaining the rod-shape (Baranowski et al. 2018). How- ral dynamics of PG synthesis in M. smegmatis and M. tuberculosis
ever, whether this serves as the sole mechanism regulating cell using super-resolution microscopy combined with fluorescent
shape is yet to be confirmed. d-alanine analogues (FDAAs) (Botella et al. 2017b). They reported
Maitra et al. 551

that FDAAs are predominantly incorporated at one of the two render the E. coli enzyme inactive (Pompeo, van Heijenoort and
poles in M. smegmatis, whereas M. tuberculosis shows variation Mengin-Lecreulx 1998; Zhang et al. 2009). The crystal structure
in polar dominance depending on the stage in cell cycle. FDAAs of M. tuberculosis GlmU reveals a unique 30-residue extension
are also incorporated along the lateral wall upon damage due to which forms a short helix at the C-terminus and is involved
muramidase activity (Garcia-Heredia et al. 2018). Furthermore, in substrate binding (Jagtap et al. 2012). The C-terminus of the
fluorescent intracellular membrane domain (IMD)-associated enzyme is also involved in auto-regulation through phospho-
protein reporters revealed that mycobacteria can spatiotempo- rylation by the serine/threonine protein kinase PknB (Parikh
rally coordinate its membrane domain in response to metabolic et al. 2009). Dziadek et al. recently reported that GlmU interacts
requirements under different growth conditions (Hayashi et al. with host immune factor IL-8, thereby increasing mycobacterial
2018). These recent reports on membrane compartmentalisation attachment to neutrophils and facilitating infection (Dziadek
of PG synthesis and its metabolism during the cell cycle in M. et al. 2016). How a cytosolic bacterial protein serves as a receptor
tuberculosis will help to understand mechanisms allowing bac- binding entity still requires clarification, however, the enzyme
teria to escape host defence systems and to characterise drug has garnered interest for drug development. The enzyme is
targeting steps that have remained elusive before. essential for M. tuberculosis and it depletion results in severe
growth defects in vitro and reduced bacillary loads in mice
models (Soni et al. 2015). Further experiments showed that the
Biosynthesis and maturation of PG
uridyl- and acetyl-transferase activities are individually essen-
The nucleotide precursors of PG were first isolated from Staphy- tial for the pathogen thus inhibitors designed for either active
lococcus aureus in 1952 (Park 1952). Since then the various steps site would be effective. Also, as the synthesis of UDP-GlcNAc
involved in the biosynthesis of PG have been extensively studied occurs via a different enzymatic route in eukaryotes compared
in a number of species. The PG biosynthetic pathway in M. tuber- to prokaryotes, any treatment designed against the latter would
culosis, however, remains largely uncharacterised. In this work be expected to have little effect on the former.
we follow the generally accepted assumption that mycobacte- MurA (UDP-N-acetylglucosamine enolpyruvyl transferase)
ria synthesise PG precursors in a similar way to the model bac- and MurB (UDP-N-acetylenolpyruvyl-glucosamine reductase)
terium E. coli using enzymes that are homologous to the ones catalyse the conversion of UDP-N-acetylglucosamine (UDP-
found in the latter. However, as a slow-growing intra-cellular GlcNAc) to UDP-N-acetylmuramate (UDP-MurNAc). MurA cataly-
pathogen with varied physiological states, mycobacteria have ses the transfer of the enolpyruvyl moiety of phosphoenolpyru-
PG enzymes that differ with respect to structure and regulation vate (PEP) to UDP-GlcNAc (De Smet et al. 1999; Xu et al. 2014).
compared to their counterparts in E. coli. This review attempts MurB (Benson et al. 1993) is an NADPH dependent oxidoreduc-
to highlight these critical features. Details of the enzymes dis- tase that reduces the enolpyruvate moiety to d-lactate result-
cussed throughout the review have been listed in Table S1 (Sup- ing in the formation of UDP-MurNAc. While mycobacteria and
porting Information). Gram-negative bacteria like E. coli have a single copy of the murA
gene (Brown et al. 1995), a second transferase gene, murA2 occurs
as a duplicate copy in low G + C Gram-positive organisms such
PG biosynthesis: the cytoplasmic steps
as S. pneumoniae (Du et al. 2000). Interestingly, both copies per-
PG biosynthesis begins with the conversion of fructose-6- form the same function and can substitute for one another. The
phosphate to UDP-N-acetylglucosamine (UDP-GlcNAc), which purpose for such redundancy at the first committed step of PG
is catalysed by the enzymes GlmS, GlmM and GlmU (van biosynthesis may be to accommodate differential regulation (Du
Heijenoort 2001a) (Fig. 2). Of these three enzymes, only et al. 2000). UDP-MurNAc binds to E. coli MurA with high affin-
GlmU has been characterised in M. tuberculosis (Zhang et al. ity, suggesting an inhibitory or regulatory role in PG biosynthesis
2009; Jagtap et al. 2012). GlmU is a bifunctional acetyltrans- (Mizyed et al. 2005). CwlM, an essential, cytoplasmic protein and
ferase/uridyltransferase, which converts glucosamine-1- hypothetical PG amidase, is another regulator of MurA (Boutte
phosphate (GlcN-1-P) to N-acetylglucosamine-1-phosphate et al. 2016). Phophorylated CwlM increases the activity of MurA
(GlcNAc-1-P) and subsequently catalyses the formation of nearly 30 fold. CwlM is phosphorylated only in the presence of
UDP-GlcNAc from GlcNAc-1-P and uridine triphosphate (Zhang surplus nutrients, thereby revealing a possible mechanism by
et al. 2009; Jagtap et al. 2012; Li et al. 2012). The reaction mech- which the pathogen correlates PG synthesis with the availabil-
anism of the acetyl transfer by GlmU provides useful insights ity of nutrients. Reducing the PG synthesis rate also has impli-
for drug design, such as the conformational changes of the cations for drug susceptibility, therefore mechanisms which can
protein structure upon interaction with the ligand (Craggs et al. activate CwlM and MurA may help to target the ‘persister’ pop-
2018). Mycobacterium tuberculosis GlmU is trimeric in solution, ulation of M. tuberculosis and reduce treatment duration, one of
whereby each monomer folds into two distinct domains. The the main causes of patient non-compliance. The regulatory role
N-terminal domain has a typical uridyltransferase fold based of CwlM has since been investigated further and its influences
on a dinucleotide-binding Rossmann fold and is similar to at later stages of PG synthesis have also been revealed and will
that observed in the reported structure for GlmU from Strep- be discussed in the following section.
tococcus pneumoniae (Sulzenbacher et al. 2001). The C-terminal Homology modelling, molecular dynamics and molecular
domain has a left-handed parallel β-helix fold forming the docking studies on the mycobacterial MurA and MurB enzymes
acetyltransferase active site containing contributions from all revealed active site residues and helped identifying potential
three subunits as is characteristic of other acetyltransferase inhibitors (Babajan et al. 2011; Kumar et al. 2011). The crystal
enzymes. However, the GlmU from M. tuberculosis is 6–8 fold structure of M. tuberculosis MurB with FAD as the prosthetic
less active than that of GlmU from E. coli. Additionally, as M. group was recently reported (Eniyan et al. 2018). MurB is a type
tuberculosis GlmU lacks free cysteines in the acetyl-CoA binding I UDP-GlcNAcEP reductase and shares conserved domains with
site or any solvent-accessible cysteines elsewhere, it retains its MurB from E. coli and Pseudomonas aeruginosa. The nicotinamide
acetyltransferase activity even in the absence of reducing agents and the enol pyruvyl moieties aligned well in M. tuberculosis
and in the presence of a thiol-reactive reagent; both of which MurB upon superimposition with the other MurB structures and
552 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

Figure 2. An overview of PG biosynthesis, degradation and recycling pathways representing information currently known in Mycobacterium species. While there has
been plethora of information on the PG metabolism in other bacteria, identification of mycobacterial proteins involved in this process has been limited so far. ‘∗’ marks
enzymes that have not been experimentally established. This includes proteins that are putative or completely unknown and uncertainty on when the enzymatic
step occurs in the pathway. The redundancy in the PG hydrolases makes it difficult to assemble a comprehensive list within a figure and only the selected enzymes
discussed in this article are highlighted. Recycling of PG in mycobacteria is an especially underexplored area compared to other bacteria and may offer interesting
insight into homeostasis mechanisms within the cell.
Maitra et al. 553

show domain III to adopt a more open conformation on bind- that screens for inhibitors of more than one enzyme in the Mur
ing to the substrates. There are known inhibitors of MurB from pathway could aid drug discovery (Basavannacharya et al. 2010a;
Gram-positive and Gram-negative bacteria, however, whether Eniyan et al. 2016).
these are active against M. tuberculosis MurB has not been inves- In addition to ATP-dependent Mur ligases, the enzymes
tigated (Bronson et al. ; Yang et al. ). catalysing the formation of substrates for Mur ligase reactions
The assembly of the peptide stem of the PG monomer unit are also key components of PG biosynthetic machinery. MurI is
(UDP-MurNGlyc or UDP-MurNAc-pentapeptide) is catalysed by a a glutamate racemase, which provides the d-Glu required for
series of four essential and structurally related ATP-dependent PG synthesis. Unlike in Bacillus subtilis there is only one gene
Mur ligases MurC, MurD, MurE and MurF which, in M. tubercu- coding for MurI in M. tuberculosis. The available crystal struc-
losis, catalyse the addition of l -Ala, d -Glu, mDAP and d -Ala- ture of MurI (Poen et al. 2016) and new inhibitors of the enzyme
d -Ala, respectively, with the concomitant hydrolysis of ATP to (Prosser et al. 2016) strengthen its potential as druggable tar-
ADP and inorganic phosphate (Fig. 2) (Basavannacharya et al. get in the pathogen. However, several issues question the value
2010a; Basavannacharya et al. 2010b; Munshi et al. 2013; Eniyan of MurI as a drug target. Homogeneous MurI purifies as a sta-
et al. 2016). The mycobacterial Mur ligases catalyse amide bond ble dimer that is enzymatically inactive in vitro. It is presumed
formation via a similar reaction mechanism to that seen in the that activator molecules such as UDP-MurNAc-l-Ala may disso-
E. coli enzymes, i.e. the formation of an activated acylphosphate ciate the dimer to generate the active enzyme as observed in
derivative of UDP-MurNAc followed by a nucleophilic attack by the case of E. coli MurI (Ho et al. 1995). This inability to extract
the amino group of the amino acid or dipeptide, ultimately and purify an enzymatically active form of the enzyme from het-
resulting in the formation of a peptide bond and release of inor- erologous expression systems poses a significant bottleneck for
ganic phosphate (Anderson et al. 1996; Falk et al. 1996; Liger et al. the development of high-throughput inhibitor screening assays.
1996; Tanner et al. 1996; Vaganay et al. 1996; Bertrand et al. 1999) Additionally, there is some ambiguity over its essentiality (Sas-
(Fig. 3E). setti, Boyd and Rubin 2003; Harth et al. 2005; Griffin et al. 2011;
All ATP dependent Mur ligases (Mur synthetases) con- Morayya et al. 2015). Mortuza et al. identified a d-amino acid
tain common structural motifs despite their low amino transaminase in M. smegmatis that could complement the activ-
acid sequence identities. ATP-dependent Mur ligases can be ity of MurI allowing murI mutants to grow in the absence of d-
described as containing three distinct domains: (i) an N- Glu (Mortuza et al. 2018). Whether its homologue in M. tuber-
terminal Rossmann-type α/β fold domain that binds to the UDP- culosis also retains d-amino acid transaminase activity is not
substrate, (ii) a central ATPase domain (with a highly conserved known. The presence of enzymes with overlapping functions
WalkerA-like motif, TG[T/S]XGK[T/S(2)]) and (iii) a C-terminal indicates that resistance would develop easily against glutamate
domain that binds the appropriate amino acid (Smith 2006) racemase inhibitors. However, as TB chemotherapy is always a
(Fig. 3). combination, the rise of resistance may be controlled by admin-
The first amino acid of the peptide stem (l-Ala) is ligated istering a combination of drugs that work synergistically, such as
onto UDP-MurNAc by MurC. In M. leprae, l-Ala is replaced by glutamate racemase inhibitors and d-cycloserine (David 2001).
Gly. Interestingly, M. tuberculosis MurC is able to incorporate both mDAP is synthesised from aspartate in eight biosynthetic
l-Ser and Gly into the growing peptide chain in vitro (Munshi steps (Usha et al. 2012). The enzymes in the pathway are essen-
et al. 2013), although it is questionable whether the UDP sub- tial and thus have the potential to become drug targets. Alpha-
strates containing these amino acids would be accepted by MurD ketopimelic acid inhibits the activity of DapA (dihydrodipicol-
in the subsequent reaction. This substrate flexibility has also inate synthase) in the micromolar range indicating that it is a
been observed in some other organisms. Addition of d-Glu by druggable target (Shrivastava et al. 2016). Mycobacterium tuber-
MurD is followed by incorporation of mDAP onto the growing culosis DapE was found to be insensitive to the known DapE
peptide chain by MurE. While over-expression of M. tuberculosis inhibitor L-captopril (Usha et al. 2016). Finally, DapF in Chlamydia
MurE in E. coli and Pseudomonas putida does not affect growth, trachomatis is bifunctional and involved in both mDAP synthesis
a lytic effect was observed when the S. aureus MurE (which and the racemisation of glutamate (Liechti et al. 2018). Whether
incorporates l-Lys instead of mDAP) was overexpressed in E. coli the DapF from M. tuberculosis could also function as a d-Glu race-
(Mengin-Lecreulx et al. 1999). In all mDAP-specific MurE ligases, mase is unknown. This essential enzyme converts ll-DAP into
an arginine residue at position 416 binds to the free end of mDAP, mDAP. Although little variation was observed in the two-domain
whereas this residue is replaced by an alanine or asparagine α/β-fold of the apo structure of DapF from M. tuberculosis, when
in all l-Lys-specific MurE ligases (Dementin 2001). MurE from compared with DapF from Haemophilus and Bacillus species, it
M. tuberculosis contains three domains, similar to that reported was noted that the M. tuberculosis DapF backbone is stabilised
for E. coli. However, unlike its E. coli homologue, the mycobacte- via a tyrosine residue specific to mycobacterial DAP epimerases.
rial MurE coordinates a magnesium ion with the UDP-MurNAc- Any change of the interactions this residue is involved in would
l-Ala-d-Glu substrate (Basavannacharya et al. 2010a; Basavan- destabilize DapF (Usha et al. 2009) thereby providing a rationale
nacharya et al. 2010b). The last two amino acids of the stem pep- for design of anti-tubercular specific DapF inhibitors.
tide are added by MurF as the dipeptide d-Ala-d-Ala Interest- The di-peptide substrate for MurF (d-Ala-d-Ala) is synthe-
ingly, MurF from E. coli accepts muropeptides with either mDAP sised by the alanine racemase Alr, followed by the d-Ala-d-Ala
or l-Lys with similar efficiencies and its N-terminus is distinct ligase Ddl. Alr is a pyridoxal-5-phosphate-containing enzyme
from the other Mur ligases (Yan et al. 2000). (van Heijenoort 2001b) and its crystal structure shows a homod-
The entire MurA-F pathway is unique to prokaryotes and imer with the domains adopting a similar fold to that observed
all corresponding genes are single-copy and essential in M. in Alr structures from other species (LeMagueres et al. 2005).
tuberculosis (Sassetti, Boyd and Rubin 2003; Griffin et al. 2011; However, the observed hinge angle between the domains is dif-
DeJesus et al. 2017) making them attractive therapeutic tar- ferent for Alr from M. tuberculosis. The interest in Alr as a drug
gets. MurC-F share conserved residues and mode of action, target has been dampened by conflicting reports on its essen-
and are thus amenable to multi-target therapy. Development tiality in M. smegmatis and M. tuberculosis. While Milligan et al.
of high-throughput activity assays as well as a one-pot assay found deletion mutants require a steady supply of d-Ala for
554 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

Figure 3. (A), Cartoon representation of the crystal structure of MurE from M. tuberculosis. The UDP substrate binding domain 1 is shown in green, the central ATPase
domain 2 is shown in blue and the amino acid substrate binding domain 3 is shown in red. Image generated using PyMOL (PDB code: 2XJA). (B), Overlay of domain
2 from MurE from E. coli (PDB code: 1E8C, light blue) and M. tuberculosis (PDB code: 2XJA, dark blue). The P-loops are highlighted in cyan. (C), Interaction map of the
UDP-MurNAc substrate with MurE from M. tuberculosis. (D), Interaction map of ADP with MurE from M. tuberculosis. (E), The reaction mechanism of Mur ligases. The
‘AA’ in the UDP-MurNAc-AA substrate represents the peptide stems of varying lengths whereas the AAx .NH2 represents the incoming amino acid.
Maitra et al. 555

growth, insertion mutants that did not show growth defects in resistance in E. coli (El Ghachi et al. 2005; Bickford and Nick 2013).
the absence of d-Ala were selected for by Marshall et al. (Milli- PgpB and BacA interact with PBP1B (which polymerises PG) and
gan et al. 2007; Awasthy et al. 2012, Marshall et al. 2017). There- stimulate its activity by dephosphorylation of the released unde-
fore, it is hypothesised that the function of Alr can be substi- caprenyl pyrophosphate (Hernández-Rocamora et al. 2018). The
tuted by an unidentified transaminase that converts pyruvate crystal structure of E. coli BacA shows a unique topology rais-
and d-Glu to d-Ala so Alr-specific inhibitors would likely have ing the possibility of its involvement as a lipid carrier flippase
no anti-mycobacterial effect. in addition to being a phosphatase that could potentially accept
Ddl catalyses the ATP-dependent ligation reaction to gener- substrates from the cytoplasm as well as the periplasm (Work-
ate the dipeptide d-Ala-d-Ala. While only one copy of ddl was man, Worrall and Strynadka 2018). Despite the redundancy of
identified in mycobacteria, two ddl genes are present in E. coli the phosphatases, deletion mutants lacking bacA show marked
(Zawadzke, Bugg and Walsh 1991). Ddl exists as a dimer, in which phenotypic effects such as attenuation of virulence in Staphylo-
each monomer contains three domains with the ligand binding coccus aureus and Streptococcus pneumoniae in mice models, and
site formed at the intersection of these three domains (Bruning impaired biofilm formation in M. smegmatis (Chalker et al. 2000;
et al. 2011). The apo form of Ddl from M. tuberculosis is unique Röse, Kaufmann and Daugelat 2004). However, the M. tuberculo-
in that in the absence of substrate it adopts a closed confor- sis homologue of BacA was found to have no role in either acid
mation that has to undergo a significant structural rearrange- resistance or in virulence in mice models (Darby et al. 2011). This
ment to bind to the substrates. The closed conformation of Ddl suggests that there may be other enzymes in M. tuberculosis with
offers many unique binding pockets adjacent to the active site decaprenyl pyrophosphate phosphatase activity. The synthesis
that may be useful for drug design. Ligand binding and catalytic and recycling of the lipid carrier is essential for the membrane-
activity, therefore, requires significantly greater structural rear- bound steps of PG synthesis as discussed below and is a verified
rangement compared to orthologues from other species such target for antibiotics such as bacitracin (Siewert and Strominger
as S. aureus. Biochemical analyses and classical enzyme kinetic 1967).
studies of Ddl from M. tuberculosis revealed the mechanism of The first membrane step in the biosynthesis of PG involves
inhibition by d-cycloserine—knowledge that could be useful in the phospho-N-acetylmuramoyl-pentapeptide transferase or
the design of future drugs (Prosser and de Carvalho 2013). Cur- translocase MraY. MraY catalyses the transfer of the phospho-
rently, d-cycloserine is known to target Ddl and Alr in M. tuber- MurNAc-pentapeptide moiety of UDP-MurNAc-pentapeptide
culosis, with strong inhibition observed in the case of Ddl and to the acceptor decaprenyl phosphate, yielding MurNAc-
weaker inhibition seen with Alr. (pentapeptide)-pyrophosphorylundecaprenol or lipid I. This pro-
ceeds via a covalent intermediate, which is formed upon nucle-
ophilic attack by an aspartate residue on the β-phosphate of
PG biosynthesis: the membrane-associated steps
the UDP-MurNAc-pentapeptide resulting in the release of UMP
A lipid carrier is essential for the transport of the hydrophilic PG (Chung et al. 2013). While E. coli uses the undecaprenyl lipid,
precursors across the hydrophobic cell membrane. Mycobacteria M. tuberculosis solely uses the decaprenyl lipid, however, both
differ from E. coli in terms of the molecule that serves as the lipid decaprenyl and heptaprenyl acceptors have been identified for
carrier (decaprenyl phosphate versus undecaprenyl-phosphate) M. smegmatis (Takayama, Schnoes and Semmler 1973). Com-
and in the synthesis of this molecule. In fact, even within prehensive biochemical or structural studies on mycobacterial
mycobacteria there are marked differences within this path- MraY have not been reported. However, active site mapping of
way. Based on sequence homology it is presumed that M. smeg- MraY from E. coli and comparison with MraY from other Gram-
matis genome codes for 4 to 10 prenyl diphosphate synthases, negative and Gram-positive bacteria revealed five conserved
whereas M. tuberculosis could contain up to 4 synthases (Crick hydrophilic sequences located on the cytoplasmic side of the
et al. 2000). Rv1086 and Rv2361 are two distinct prenyl diphos- membrane containing invariant amino acids (Al-Dabbagh et al.
phate synthases, a ω,E,Z-farnesyl diphosphate synthase (Z- 2008). An X-ray crystal structure for MraY from Aquifex aeolicus
FPPS) and a decaprenyl diphosphate (DecaPP) synthase, respec- and a method to quantify MraY activity has provided further
tively (Schulbach, Brennan and Crick 2000). These act sequen- insight into the active site residues (Chung et al. 2013). Improve-
tially to synthesise decaprenyl diphosphate from isopentenyl ments of the biochemical assay to quantify MraY activity (Siri-
diphosphate and E-geranyl diphosphate (Schulbach, Brennan cilla et al. 2014) is a significant asset in the effort to develop
and Crick 2000; Schulbach et al. 2001; Kaur, Brennan and Crick antimycobacterial agents targeting this protein.
2004). Rv1086 adds one isoprene unit to geranyl diphosphate, Following the synthesis of lipid I, the N-acetylglucosamine
generating the 15-carbon product (E,Z-farnesyl diphosphate). transferase MurG catalyses the transfer of GlcNAc from UDP-
Rv2361c then processively adds a further seven isoprene units GlcNAc to lipid I, yielding GlcNAc-MurNAc-(pentapeptide)-
to E,Z-farnesyl diphosphate to generate the 50-carbon prenyl pyrophosphoryl-decaprenol or lipid II.
diphosphate. The crystal structures of the enzymes show that MurT and GatD are responsible for the amidation of the d-
they are closely related, which partly explains how Rv2361 can Glu residue in the peptide stem of both, lipid I and lipid II in
compensate for the deletion of Rv1086 (Schulbach et al. 2001; Staphylococcus aureus and Streptococcus pneumoniae (Figueiredo
Wang et al. 2008). The decaprenyl diphosphate, synthesised de et al. 2012; Munch et al. 2012). The arrangement of the murT and
novo at the cytoplasmic side of the cell membrane or released by gatD genes in an operon is conserved across various bacterial
the polymerisation of PG on the periplasmic face of the mem- families (Figueiredo et al. 2012) and has also been confirmed in
brane, must be dephosphorylated for the transfer of the acti- mycobacteria (Maitra et al., unpublished results). Crystal struc-
vated sugar molecule from lipid I. The dephosphorylation of tures of the enzymes from S. aureus and S. pneumoniae reveal that
decaprenyl diphosphate to decaprenyl phosphate is performed the pair form a stable, functional heterodimer wherein GatD is
by membrane-bound pyrophosphatases. In spite of multiple responsible for the sequestration of the glutamine donor and its
undecaprenyl pyrophosphate phosphatases (Upp) identified in subsequent deamination and MurT is a Mur ligase-like protein
E. coli (BacA, PgpB, YbjG and LpxT) the majority of this activity that allows for the d-iGlu in the stem peptide to be amidated
(75%) is attributed to BacA (UppP); which also confers bacitracin to d-iGln and utilises an ATP in the process (Leisico et al. 2018;
556 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

Morlot et al. 2018; Noldeke et al. 2018). While GatD is inactive in have GTase activity, but only when in complex with its cognate
the absence of MurT, the latter can functionally substitute the penicillin-binding protein (Taguchi et al. 2019).
role of MurE (Munch et al. 2012; Leisico et al. 2018). Amidation of The initial hypothesis for MurJ as the lipid II flippase was
PG plays a crucial role in PG synthesis and maturation. It was based on its essentiality and the observation that PG synthe-
hypothesised that amidation could facilitate the translocation sis stops and precursors accumulate upon depletion of the murJ
of lipid II across the cytoplasmic membrane as a consequence gene in E. coli (Inoue et al. 2008; Ruiz 2008). A colicin M-based
of the reduction of polarity (Munch et al. 2012). It has also been cellular assay suggested that MurJ might flip lipid II across the
reported that non-amidated lipid II is an inefficient substrate for membrane (Sham et al. 2014). Native mass spectroscopy showed
transpeptidation reactions in S. pneumoniae (Zapun et al. 2013). that MurJ binds lipid II and that this binding is affected by car-
Amidated muropeptides are recognized by PknB in M. tubercu- diolipin (Bolla et al. 2018). The crystal structures of MurJ from
losis and through the action of the kinase could regulate cell Thermosipho afticanus (Kuk, Mashalidis and Lee 2017) and E. coli
growth, division and PG turnover (Mir et al. 2011). (Zheng et al. 2018) confirmed similarity to MOP family of trans-
The homologous murT/gatD genes in M. tuberculosis are porters and further rationalised possible mechanisms by which
essential for in vitro growth of the pathogen (Sassetti, Boyd and MurJ is likely to transport lipid II across the membrane. However,
Rubin 2003). The essentiality of MurT/GatD and the requirement until there is direct evidence with the mycobacterial protein, the
for complex formation for activity may be exploited for design- identity of the flippase in the pathogen remains uncertain.
ing drug scaffolds. Both PG precursor synthesis and their transport are regu-
Mycobacterial lipid II also contains amidated mDAP residues lated by PknB-mediated phosphorylation of CwlM (introduced
(Mahapatra et al. 2005b). In Corynebacterium glutamicum and B. earlier as regulating MurA activity). CwlM exists in two forms
subtilis, ltsA and asnB encode for the amidotransferases respon- within the mycobacterial cell. The non-phosphorylated CwlM
sible for the amidation of mDAP (Levefaudes et al. 2015; Dajkovic is membrane-associated, interacts with MurJ and presumably
et al. 2017). LtsA and AsnB belong to the growing family of glu- enables it to transport lipid II (Turapov et al. 2018). The phos-
tamine amidotransferases (such as GatD) whose members catal- phorylated CwlM is cytoplasmic and interacts with FhaA and
yse amide nitrogen transfer from glutamine to various spe- MurA. The authors hypothesise that on binding to unpoly-
cific acceptor substrates (mDAP in this case). Most of the inves- merised PG precursors in the periplasmic region, PknB under-
tigations into the role of mDAP amidation have focussed on goes autophosphorylation and in turn phophorylates its sub-
the host immune response modulation of such modification. strate CwlM, which dissociates from MurJ thereby reducing the
Nucleotide-binding oligomerisation domain-containing protein flipping of PG precursors. Direct phosphorylation of MurJ by
1 (NOD1) is a pattern recognition receptor that recognizes PknB was also proposed to produce the same outcome.
MurNAc-tripeptide fragments containing mDAP and activates A number of compound classes bind to extracytoplasmic
the innate immune system (Girardin et al. 2003b). Amidation lipid II and prevent PG polymerisation. Glycopeptides such as
of mDAP enables evasion of recognition by NOD1 by interact- vancomycin (MICMtb - 65 μg/mL) bind to d-Ala-d-Ala in lipid II
ing with residues away from the NOD1-leucine-rich repeat (LRR) and nascent PG. Antimicrobial peptides such as nisin (MICMtb -
recognition region (Roychowdhury, Wolfert and Boons 2005; > 60 μg/mL), teixobactin (MICMtb - 0.125 μg/mL) and malacidin
Wolfert, Roychowdhury and Boons 2007; Vijayrajratnam et al. (MICMtb - not determined) bind primarily to the pyrophosphate
2016; Wang et al. 2016). Unlike d-Glu amidation, the modifica- moiety of lipid II and inhibit PG synthesis.
tion of mDAP residue does not influence the pattern of insertion
of PG precursors into the cell wall or the overall cross-linking
PG biosynthesis: linkage in mature PG
levels (Levefaudes et al. 2015; Dajkovic et al. 2017). However, the
amidation of the mDAP residues in mycobacteria is essential for Once the precursors are translocated to the periplasmic space,
the formation of LD-cross-links (Ngadjeua et al. 2018). Addition- lipid II is used as the substrate for the subsequent polymerisa-
ally, the presence of zones of uncontrolled PG degradation in tion reactions and the formation of mature PG. The final stages
the asnB mutant of B. subtilis suggests that mDAP amidation of PG maturation occurs in the periplasm, carried out by trans-
controls the activity of hydrolytic enzymes such as autolysins glycosylases and transpeptidases, which are often bifunctional
(Dajkovic et al. 2017). Once the modifications have been intro- penicilloyl serine transferases or penicillin binding proteins
duced, lipid II is flipped to the outer face of the cytoplasmic (PBPs) (Crick, Mahapatra and Brennan 2001; Sauvage et al. 2008),
membrane. resulting in the formation mature of PG chains and peptide with
The translocation of lipid II across the cytoplasmic mem- DD-cross-links (Goffin and Ghuysen 1998). The class A and B
brane has been subject to some controversy, with the role of PBPs synthesize PG; whereas the class C PBPs are PG hydrolases
lipid II flippase having being contested between FtsW and MurJ. with either DD-endopeptidase or DD-carboxypeptidase activ-
The evidence supporting FtsW as the lipid II flippase comes ity. Both reactions provide the tetrapeptide substrates for LD-
from an assay performed by Mohammadi et al. (Mohammadi transpeptidases. Class A PBPs (in mycobacteria PBP1 and PBP1A,
et al. 2011) on an in vitro reconstitution system, in which it was encoded by ponA1 and ponA2, respectively) consist of bifunc-
observed that FtsW, and not MurJ, was able to translocate flu- tional enzymes with an N-terminal glycosyltransferase and a C-
orescently labelled lipid II across the membrane of bacterial terminal DD-transpeptidase (penicillin-binding) domain (Maha-
membrane vesicles. It was later suggested by the same research patra et al. 2000; Bhakta and Basu 2002; Patru and Pavelka
group that lipid II may be translocated via a pore-like structure 2010). Class B PBPs (in mycobacteria PbpA and PbpB) are mono-
in FtsW (Mohammadi et al. 2014). Conversely, it has been sug- functional DD-transpeptidases (Dasgupta et al. 2006; Plocin-
gested based on genetic analyses and biochemical assays with ski et al. 2011). The glycosyltransferase domain catalyses the
RodA, that proteins from the SEDS family may be glycosyltran- transfer of the terminal N-glycolylmuramic acid residue from a
ferases (GTases) rather than lipid II flippases (Meeske et al. 2016; nascent glycan chain to N-acetylglucosamine of lipid II, releas-
Emami et al. 2017). However, FtsW was not active as GTase in ing decaprenol pyrophosphate (Scheffers and Pinho 2005; van
another study in which it was also observed that FtsW, but not Heijenoort 2007). The C-terminal DD-transpeptidase domain
MurJ, binds lipid (Leclercq et al. 2017). However, FtsW does in fact of PonA1 crosslinks the penultimate d-Ala to the mDAP of
Maitra et al. 557

another peptide (Filippova et al. 2016). The energy for the DD- peptide cross-links in the absence of de novo synthesis. These
transpeptidation reaction stems from the cleavage of the d-Ala- features might facilitate the repair of damaged PG (Morè et al.
d-Ala peptide bond of the donor peptide resulting in the release 2019) or render PG resistant to hydrolytic enzymes (Lavollay et al.
of the terminal d-Ala residue (Ghuysen 1991). PonA localises to 2008), which could be vital during stationary phase or persis-
the poles and septum in mycobacteria and is essential in reg- tence.
ulating cell length probably through its interaction with both PG transpeptidases are verified but difficult targets for
PknB and the peptidoglycan hydrolase RipA (Hett, Chao and antimicrobial drug discovery as (i) due to their redundancy,
Rubin 2010; Kieser and Rubin 2014). The activities of PonA1 many of these enzymes can substitute the function of another
and PonA2 may be altered under different growth conditions as and (ii) the drugs most commonly used to target them, the β-
mutants lacking one of the corresponding genes show differen- lactams, are prone to inactivation by β-lactamases.
tial response to cell wall antibiotics (Kieser et al. 2015). Drug development efforts targeting the LD-transpeptidases
The LD-cross-link was first identified by Wietzerbin in 1974 have primarily focussed on further exploring the potential
(Wietzerbin et al. 1974). Its physiological significance, however, of carbapenems, particularly meropenem, as Ldt inhibitors.
was revealed only in 2008 when Lavollay and colleagues dis- Shortly after the identification of LdtMt1 , Hugonnet and col-
covered that the PG of stationary phase M. tuberculosis almost leagues demonstrated that a combination of meropenem and
exclusively contained LD-cross-links (Lavollay et al. 2008). An clavulanate was able to kill 13 different XDR strains of M.
LD-transpeptidase, LdtMt1 , which catalysed the formation of LD- tuberculosis with MIC values > 1 μg/mL (with one exception at
cross-links, was identified and covalent adduct formation with 1.25 μg/mL) (Hugonnet et al. 2009). This built upon previous
the β-lactamase drugs imipenem, meropenem and ertapenem work demonstrating that clavulanate can act as an irreversible
was demonstrated through mass spectroscopy (Lavollay et al. inhibitor of the M. tuberculosis β-lactamase BlaC (Hugonnet and
2008). Subsequently, a second LD-transpeptidase, LdtMt2 was Blanchard 2007).
identified by Gupta and colleagues, and loss of the ldtMt2 gene in The direct targeting of LdtMt1 by carbapenems and
M. tuberculosis was shown to result in altered colony morphol- cephalosporins was investigated by Dubee and colleagues
ogy, loss of virulence and increased sensitivity to amoxicillin- and it was demonstrated that while both classes of β-lactamase
clavulanate (Gupta et al. 2010). In addition, LdtMt2 is critical for are able to form adducts with LdtMt1 , cephalosporins do so 7-
growth in the absence of PonA1 or PonA2, suggesting PBPs and to 1000-times slower than carbapenems (Dubee et al. 2012).
LdtMt2 together support new cell wall synthesis during growth Since then, Kim et al. showed that meropenem acts as a suicide
(Kieser et al. 2015). inhibitor of LdtMt2 by inducing conformational changes in the
The influence of LD-cross-linking on mycobacterial phys- protein; further informing drug design groups about inhibition
iology and drug susceptibility was further investigated by mechanisms for the protein (Kim et al. 2013).
Baranowski and colleagues, making use of an M. smegmatis Recently, in a small retrospective observational case study,
strain lacking Ldts as a model (Baranowski et al. 2018). It was Paven and colleagues reported that of 18 patients with XDR-
observed that in the absence of LD-cross-links, the rigidity of TB who were treated with meropenem-clavulanate, 15 had
aging cell wall was compromised leading to the formation of a ‘successful outcome’ (5 ‘cured’ and 10 ‘completed treat-
spherical blebs. The increased susceptibility of M. tuberculosis ment’), while two patients failed to complete the treatment
to DD-transpeptidase targeting β-lactams was also confirmed and one died (Payen et al. 2018). It is also notable that no
and it was demonstrated that inhibiting both DD- and LD- adverse effects were observed as a result of the treatment, sug-
transpeptidases with amoxicillin and meropenem respectively, gesting that with further study this may present a promis-
resulted in synergistic lowering of the MIC (Baranowski et al. ing route to new β-lactamase-based TB treatment. However,
2018). treatment of Mycobacterium bovis BCG with meropenem was
The two-step reaction of LD-transpeptidases begins with the also reported to cause a significant increase in endogenous
acylation of the enzyme by the third residue (l-chiral centre) ATP levels and suppression of this ATP spike using drugs
of the tetrapeptide donor, followed by deacylation of this acyl- such as Bedaquiline attenuated the activity of the former
enzyme intermediate by the third residue (d-chiral centre) of (Shetty and Dick 2018). Therefore, the inclusion of new drugs
the adjacent acceptor stem resulting in the formation of a LD- into combination therapy poses complications of antagonistic
peptide linkage. The crystal structure of the extra-membrane activity.
domain of LdtMt2 with a bound PG fragment (Erdemli et al. 2012)
showed that each monomer consists of two globular domains:
PG biosynthesis: orientation of mature PG
an N-terminal domain resembling an immunoglobulin domain,
and a C-terminal catalytic domain, consisting of a β sand- The orientation of PG has been a subject of debate and specu-
wich with two mixed β sheets characteristic of the YkuD fold lation throughout the past decades. The classical depiction rep-
(Bielnicki et al. 2006), with the two domains connected by a resents PG glycan strands as parallel to the bacterial membrane
short linker molecule. Structural, kinetic and calorimetric data surface as this provides better flexibility to accommodate cell
revealed a catalytic mechanism for LdtMt2 in which both acyl- division (Koch 1998, Koch 2000; Vollmer and Holtje 2004; Schef-
acceptor and acyl-donor substrates enter from the same side to fers and Pinho 2005; Vollmer 2008), however, others postulate
reach the catalytic site (Both et al. 2013; Triboulet et al. 2015; Bhat- that PG is arranged perpendicular/orthogonal to the membrane
tacharjee et al. 2019). (Pink et al. 2000; Dmitriev et al. 2003; Dmitriev, Toukach and
The PG layer was long regarded as a network with mainly DD- Ehlers 2005; Meroueh et al. 2006). Using atomic force microscopy
cross-links. However, the type of cross-links in PG can switch the glycan strands of E. coli were seen running approximately
to mainly LD-cross-links in response to changes in the environ- along the short axis of the cell, and the PG was more ordered
ment and growth phase in Mycobacteria (Gupta et al. 2010) and in rod shaped E. coli sacculi than spheroid shaped sacculi from
E. coli (Hugonnet et al. 2016). The shift to LD-cross-links allows mutant strains (Turner et al. 2018). Our current understanding of
cells to grow in the presence of most β-lactam antibiotics that the order of PG glycan strands is largely derived from studies on
target PBPs and not LD-transpeptidases, and the use of tetrapep- rod-shaped E. coli and B. subtilis, where new glycan strands are
tide donors by LD-transpeptidases allows for the generation of inserted circumferentially forming new PG laterally along the
558 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

side wall with the help of a protein complex called elongasome of PG, while N-acetylmuramyl-l-Ala amidases hydrolyse the
(Dominguez-Escobar et al. 2011; Garner et al. 2011; Hussain et al. amide bond between MurNAc and l-Ala of the stem peptide.
2018). Whether a similar order of PG glycan strands is present in Lytic transglycosylases are, by virtue of their enzymatic reac-
mycobacteria is not known, as there are key differences between tion, not ‘hydrolases’ but they are usually discussed amongst
mycobacterial PG biosynthesis and other rod-shaped bacterial the PG hydrolases. Unlike other N-acetyl-β-d-muramidases like
PG structure such as the incorporation of nascent PG at polar lysozyme the lytic transglycosylases catalyse the formation of
ends, the presence of a high proportion of LD-cross-linkages and a 1,6-anhydromuramyl ring by an intramolecular transglycosy-
the absence of MreB. The complexity of the mycobacterial cell lation reaction in the MurNAc residue of the released PG frag-
wall structure, lack of pure samples and the presence of abun- ment. Different forms of endopeptidases and carboxypeptidases
dant secondary cell wall polymers make it challenging to deter- are capable of hydrolyzing amide bonds either in the stem pep-
mine its architecture. tide or cross-linkages of PG (Scheurwater, Reid and Clarke 2008;
Vollmer et al. 2008).
The presence of three major autolysins, N-MurGly-l-Ala ami-
PG biosynthesis: the dcw gene cluster
dase, an amino peptidase and an endopeptidase in M. smegmatis,
Although most of the genes involved in PG biosynthesis have was first reported in 1977 (Kilburn and Best 1977) followed later
been identified in E. coli and their mycobacterial orthologues by partial characterisation of a β-glycosidase from M. phlei (Li
are known, the differences in the structure of PG between et al. 1999).
the organisms, however, warrants further investigation into DacB2 from M. tuberculosis is a DD-carboxypeptidase (Class
the products of these key genes. Since the sequencing the C PBP) that removes d-Ala residues at position 5 of pentapep-
genome of M. tuberculosis (Cole et al. 1998), a plethora of infor- tides (Kumar et al. 2012). The overexpression of dacB2 results in
mation is available about the pathogen, aiding a more targeted altered morphology and defective biofilm formation, while its
approach towards drug discovery and development. The major- deletion leads to increased intracellular survival of the mutant
ity of PG biosynthetic genes in M. tuberculosis are located on in THP-1 cells (Bourai, Jacobs and Narayanan 2012). DacB2 was
the division-cell wall (dcw) cluster along with genes required also reported to possess DD-endopeptidase activity (Baranowski
for cell division (Munshi et al. 2013). This clustering of genes et al. 2018). Thus, it can affect cell morphology by hydrolysing
is also observed in E. coli (Eveland, Pompliano and Anderson the DD-cross-links as well providing tetrapeptide substrates for
1997), with the exception of the Rv2161c-Rv2160c-Rv2159c region LD-transpeptidases by removing the terminal d-Ala residue.
and the homologues present only in the slow growing mycobac- Early investigations by Votyakova (Votyakova et al. 1994) and
terial species such as M. tuberculosis (Fig. 4). The clustering Mukamolova (Mukamolova et al. 1998b; Mukamolova et al. 1999;
of cell division genes (ftsZ, ftsQ and ftsW) and PG biosynthe- Mukamolova et al. 2002) in Micrococcus luteus suggested the pres-
sis genes in the dcw operon are indicative of a complex and ence of a resuscitation promoting factor (Rpf) with lytic transg-
spatio-temporal coordination between cell division proteins and lycosylase activity responsible for the release of PG fragments
cell wall precursor synthesis enzymes (Mingorance, Tamames (Fig. 2). This ground-breaking discovery led to the identifica-
and Vicente 2004; Vicente et al. 2006; Munshi et al. 2013; Carel tion of five Rpfs (rpfA-rpfE) in Mycobacterium spp. (Cole et al.
et al. 2014). 1998; Mukamolova et al. 1998a; Sutcliffe and Harrington 2004),
which significantly enhanced our understanding not only of
the remodelling of cell wall PG during daughter cell formation,
Degradation and remodelling of PG
but also the processes behind dormancy and reactivation of
PG is a dynamic structure that is constantly synthesised and bacilli during active and latent TB. Although all five Rpfs are
remodelled. Cell growth, division and septation require the dispensable for mycobacterial growth, the lack of three of the
co-ordination of the enzymes required for both PG synthe- five rpfs leads to avirulence in mice and an inability to resus-
sis and hydrolysis. PG hydrolases (PGH) form a vast group citate from dormant bacilli (Downing et al. 2005). Furthermore,
of enzymes including glycosidases, amidases, endopeptidases the lack of spontaneous reactivation of the rpf deletion mutants
and carboxypeptidases, which together are capable of fully reveals a synergy between them, suggesting that their func-
digesting the PG polymer into soluble fragments. They can act tion cannot be substituted by another enzyme (Downing et al.
by cleaving bonds within polymeric or soluble fragments of 2005, Kana et al. 2008). Interestingly, an Rpf-related motif iden-
PG (Loessner et al. 2002; Vollmer et al. 2008), and hence may tified in mycobacteriophages is involved in PG degradation to
have a periplasmic, membrane-associated or cytoplasmic loca- facilitate phage infection during the stationary phase (Piuri and
tion (Holtje 1998). Often called autolysins, they are capable of Hatfull 2006). This suggested that the Rpf-related motif was
lysing whole cells when deregulated. Due to their selective important for the breakdown of PG with extensive ld-cross-links
ability to cleave PG, the hydrolases have been implicated in a such as those encountered in dormant M. tuberculosis. The Rpfs
plethora of cellular processes including cell growth, PG matu- have detectable sequence homology with lysozymes and lytic
ration, PG turnover, cell division, sporulation, resuscitation and transglycosylases (Cohen-Gonsaud et al. 2004; Mukamolova et al.
pathogenicity (Goodell and Schwarz 1985; Heidrich et al. 2002; 2006). The C-terminal domain of RfpB from M. tuberculosis com-
Morlot et al. 2010; Lee and Huang 2013). Due to their central role prises a PG binding cleft with two parts. One forms a compact
in the processes outlined above these enzymes offer a unique lysozyme-like fold resembling that of c-lysozyme, whereas the
opportunity to alter the permeability of the mycobacterial cell other resembles soluble lytic transglycosylase proteins such as
wall and so make M. tuberculosis and related pathogens more E. coli Slt70 (van Asselt, Thunnissen and Dijkstra 1999; Cohen-
susceptible to drug treatment. In support of their importance Gonsaud et al. 2005). Although the catalytic domains in RpfE and
in regulation of bacterial physiology Machowski et al. recently RpfC are similar to those in RfpB (Chauviac et al. 2014; Mavrici,
identified multiple homologues of these PG remodelling genes Prigozhin and Alber 2014), the substrate-binding site in RpfE is
in M. tuberculosis (Machowski et al. 2014). more positively charged than that in RpfB. This suggests that
Glycosidases such as N-acetyl-β-d-glucosaminidases and N- these Rpfs either function optimally at different pH values or
acetyl-β-d-muramidases cleave the polysaccharide backbone cleave different micro-domains of PG.
Maitra et al. 559

Figure 4. Conservation in genomic organization of dcw gene clusters. Here, we compare the differences in the gene content of the dcw clusters of fast and slow growing
mycobacteria with that of E. coli.

Due to the network-like structure of PG, its enlargement at differing growth rates. RipB has unique features in its N-
during cell growth and division requires the synergistic activ- terminal segment, which runs across the catalytic core domain
ity of both biosynthetic and hydrolytic enzymes for the incor- and forms a helix instead of the β-hairpin loop seen in the
poration of new glycan strands into the growing PG sacculus. homologous module of RipA (Both, Schneider and Schnell 2011).
PG hydrolases can destroy the integrity of the sacculus; hence These enzymes are not essential and so are not attractive drug
their activity must be controlled in the cell to avoid autolysis. targets.
There is mounting evidence that the biosynthetic and hydrolytic N-acetylmuramyl-l-Ala amidases hydrolyse the bond
enzymes interact with each other to form complexes capable between MurNAc and the peptide portion of the PG and have
of synergistic breaking and making of bonds in PG and regu- been implicated in PG degradation and turnover, cell separation,
lating activity. In E. coli, Slt70 (a soluble lytic transglycosylase) antibiotic resistance and spore formation (Heidrich et al. 2001;
forms a complex with PBP 3 (a PG transpeptidase) and PBP 7/8 Vollmer et al. 2008). PG amidases fall into two groups; one
(a DD-endopeptidase) and both enzymes synergistically degrade containing the amidase 2 domain and other the amidase 3
PG (Romeis and Holtje 1994). Slt70 also interacts with PBP1B, zinc-dependent domain (which includes the E. coli AmiA, AmiB
PBP1C and PBP 2 (von Rechenberg, Ursinus and Holtje 1996). The and AmiC and B. subtilis CwlB and CwlC enzymes). The first
lytic transglycosylase MltA interacts with PBP1B (a bifunctional autolysin identified in M. tuberculosis was an amidase known
synthase) and MipA (MltA-interacting structural protein). MtlB as CwlM (Rv3915), which has since been established to have a
interacts with PBP1B, PBP1C and PBP 3 (von Rechenberg, Ursi- regulatory role in PG synthesis and maintenance as mentioned
nus and Holtje 1996; Vollmer, von Rechenberg and Holtje 1999). in preceding sections. Another homologue (Rv3717) of the
In Mycobacterium spp. the resuscitation promoting factors RpfB E. coli PG amidases recently identified in M. tuberculosis was
and RpfE interact with the endopeptidase RipA at the septum of found to lack a PG-binding domain and is active on muramyl
dividing bacteria (Hett et al. 2007; Hett and Rubin 2008). An inter- dipeptide, but not on polymerised PG (Kumar et al. 2013;
action between SltB1 and PBP2 has been observed in P. aeruginosa Prigozhin et al. 2013). The catalytic function identified by the
(Legaree and Clarke 2008; Nikolaidis et al. 2012). structural analysis of Rv3717 indicates that the catalytic pocket
Mycobacterial RipA is a d-Glu-mDAP endopeptidase that of this enzyme is shaped as a blind tunnel and accommodates
cleaves between positions 2 and 3 of the stem peptide (Both, only PG monomers, as peptide cross-links in polymerised PG
Schneider and Schnell 2011). RipA is essential in M. tuberculosis cannot enter the tunnel. It uses an overall positive charge on its
(Sassetti, Boyd and Rubin 2003) and its depletion in M. smegmatis surface to bind to its substrate and regulates its activity through
causes severe growth inhibition (Hett and Rubin 2008). Unregu- an auto-regulatory β-hairpin (Kumar et al. 2013). As this is a
lated, RipA behaves like an autolysin in M. tuberculosis and M. cytosolic protein the activity of this enzyme depends on the
smegmatis, resulting in spherical cells that eventually lyse (Chao action of another PG hydrolase cleaving the cross-links of PG
et al. 2013). RipA, belonging to NlpC/P60 family, has a Cys-His- fragments released from the cell wall and transported back into
Glu catalytic triad residing in the C-terminal catalytic domain, the cytoplasm. This indicates a possible involvement for Rv3717
(Ruggiero et al. 2010) and is regulated by proteolytic cleavage by in PG recycling (Prigozhin et al. 2013).
MarP (Botella et al. 2017a). RipA becomes a lethal autolysin in
the absence of protein interactions that are necessary to con- Transport and recycling of degraded PG components
trol its proteolytic activation (Chao et al. 2013). RipA-mediated A study of turnover rates in E. coli led to the serendipitous dis-
cleavage in PG and its activation is more robust in the fast- covery of PG recycling (Goodell and Schwarz 1985). The extent of
growing environmental species M. smegmatis compared to the PG fragments released from the cell wall of growing bacteria, is
slow growing M. tuberculosis or M. bovis BCG—presumably a con- in the range of 25%–50% in Gram-positive organisms such as B.
sequence of the varying demands for PG hydrolysis required
560 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

subtilis (Mauck, Chan and Glaser 1971) and E. coli (Chaloupka and NagA and indicates that the active site does not undergo any
Strnadova 1972; Goodell 1985; Goodell and Schwarz 1985). How- major structural change on binding the substrate. However, two
ever, E. coli recycles about 90% of the turnover products, releas- loop regions demonstrate a capacity for conformational flexibil-
ing only 0%–5% of the PG material into the culture supernatant. ity and an importance in facilitation of substrate binding, pro-
Until recently, it was believed that PG recycling is a characteris- viding opportunity for designing inhibitor molecules (Ahangar
tic of Gram-negative bacteria only (Herve et al. 2007). However, et al. 2018). Biochemical characterization shows a clear prefer-
the identification of orthologues of recycling enzymes in B. sub- ence of NagA for GlcNAc6P over other amino sugar analogues.
tilis and subsequent muropeptide recovery studies have uncov- The stringent recognition of this fragment is hypothesised to
ered evidence of PG recycling in Gram-positive organisms as well ensure the integrity of PG recycling (Ahangar et al. 2018).
(Reith and Mayer 2011; Kluj et al. 2018). Apart from the obvi- The essentiality of nagA makes it a feasible drug target.
ous advantages associated with conservation of resources and Eukaryotic homologues of the enzyme have low sequence iden-
energy, PG recycling also plays a significant role in monitoring tity and accommodate larger, glycolyl substituted substrates.
the integrity of the cell wall, cell signalling and antibiotic resis- This property makes the two homologues distinct, however,
tance via β-lactamase induction (Jacobs et al. 1994). Additionally small inhibitors acting on M. tuberculosis NagA may be accepted
PG recycling plays a crucial role in the suppression of the innate by mammalian homologues thereby giving rise to cytotoxicity
immune response, by effectively recovering cell wall muropep- (Bergfeld et al. 2012).
tides that would otherwise stimulate immunity, via proteins that Recycling of stem peptides is facilitated by murein peptide
recognise PG (Boudreau, Fisher and Mobashery 2012; Johnson, ligase (Mpl) in Gram-negative organisms. Mpl ligates di-, tri- and
Fisher and Mobashery 2013). tetrapeptide stems onto UDP-MurNAc in an ATP-dependent step
Although there is little knowledge about PG recycling in thereby replacing the functions of MurC-F playing an impor-
M. tuberculosis, some similarities have been observed with tant role in the transition of cell towards stationary phase, as
the well-studied system in E. coli. Recycling of PG precursors observed in E. coli (Talukder et al. 1996). An mpl homologue has
entails lateral wall and septal PG degradation to form GlcNAc- not been identified in Gram positive bacteria or in any mycobac-
(1→4)-1,6-anhydro-MurNAc-peptide monomers, GlcNAc-(1→4)- terial species.
1,6-anhydro-MurNAc dissacharides and free peptides (Goodell
1985). Pulse-chase experiments with tritiated mDAP provided
Role of PG in cell signalling
evidence to suggest that many of the cell wall precursors were
recycled (Goodell and Schwarz 1985). AmpG from E. coli, also PG fragments released from the cell envelope during growth
required to induce AmpC (a β-lactamase), was identified as and cell division play important roles in cellular signalling and
the permease transporting the primary products of the action inter-bacterial communication (Humann and Lenz 2009) (Fig. 5).
of lytic transglycosylases on PG into the cytoplasm (Korfmann Rpfs produce PG fragments that lead to the resuscitation of dor-
and Sanders 1989; Jacobs et al. 1994; Cheng and Park 2002). mant mycobacteria (Mukamolova et al. 1998a; Mukamolova et al.
Although Gram-positive organisms lack an AmpG homologue, 1999; Nikitushkin et al. 2013). RpfC and RpfD are restricted to
the presence of muropeptides with various length peptide side pathogenic and environmental mycobacteria and are secreted
chains (tetrapeptides, dipeptides) (Mahapatra et al. 2008), as well into the culture medium, unlike RpfA, RpfB and RpfE, which
as inducible β-lactamases (Flores, Parsons and Pavelka 2005), are membrane-bound (Machowski et al. 2014). This implies that
suggests that an equivalent pathway for the recycling path- specific PG products are likely to act as messengers, which can
way for PG exists in mycobacteria. Once the muropeptides have act as a signal to reawaken dormant cells (Mukamolova et al.
been transported back to the cytoplasm, various amidases and 1998a; Zhang et al. 2001; Machowski et al. 2014). The release of
epimerases, which are non-essential for growth, further process mDAP-containing muropeptides during growth triggers sporula-
them to products that can be utilised for de novo PG precursor tion in B. subtilis and by binding of the serine threonine protein
synthesis (Park and Uehara 2008). kinase (STPK) PrkC to the PG fragment (Shah et al. 2008). It is
Homologues of the enzymes involved in the import, break- possible that in mycobacteria the PG fragments released by Rpfs
down and recycling of muropeptide in the cytoplasm are yet to bind PknB, triggering a cascade of signals leading to resuscita-
be identified in M. tuberculosis. In the preceding section we dis- tion of the bacteria (Fig. 5). Furthermore, Russell-Golman et al.
cussed the various enzymes involved in degradation and remod- also showed that mycobacterial double mutants of RpfA and
elling of the cell wall PG. Recent work by Moynihan et al. pro- RpfE show a reactivation-deficient and attenuated phenotype
vided evidence for the uptake of muropeptide fragments in M. (Russell-Goldman et al. 2008), suggesting that Rpfs serve as vir-
tuberculosis and M. bovis BCG; where they show cleavage of stem ulence factors (Romano et al. 2012). In addition, Rpfs induce ele-
peptide followed by disaccharide cleavage by LpqI (NagZ) and vated levels of cytokines in cultures from individuals with latent
finally lactyl-ether removal from the MurNAc moiety (Moyni- TB (Riano et al. 2012).
han et al. 2019). NagA is involved in PG recycling through the Although PG has been the subject of intense study over the
deacetylation of GlcNAc-6P in a number of other organisms, last number of decades, the changes that occur when a cell
including E. coli, B. subtilis, S. aureus and Streptomyces coelicolor. enters and exits stationary phase are not well understood. In
A putative nagA gene in an operon distinct from other bacterial some bacteria, d-amino acids are incorporated into the PG poly-
organisms, has been identified in M. tuberculosis (Ahangar et al. mer in the stationary phase, which affects the amount of PG per
2018). Mycobacterium tuberculosis nagA is essential and encoded cell (Lam et al. 2009; Cava et al. 2011). d-Ala inhibits the germi-
along with genes involved in PG biosynthesis and carbohydrate nation of spores in B. subtilis (Chesnokova et al. 2009). d-Met and
uptake, indicating that it may play a role in the recycling of d-Leu were the most prominent non-canonical d-amino acids
GlcNAc6P derived from the breakdown of PG. incorporated into the PG during the stationary phase in Vibrio
Structural characterisation of NagA reveal it to be a dimer, cholerae (Lam et al. 2009).
with each unit organised in two domains with active site pockets The ability of a host to recognise bacterial components
occurring in domain I (Ahangar et al. 2018). The crystal structure and trigger an innate immune response is the first line of
reveals the binding of the GlcNAc-6P substrate to M. smegmatis defence against infection. The fact that eukaryotes do not
Maitra et al. 561

Figure 5. The role of PG and its breakdown products in signalling. The digestion of PG by hydrolases generates products like muramyl dipeptide and muramyl tripeptide
that are key signalling molecules to resuscitate bacterial cells and may induce β-lactamase. PG fragments are encountered by the host immune surveillance systems
either extracellularly (e.g. in serum) or are phagocytosed triggering various pro-inflammatory host immune responses.
562 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

produce PG has led to the evolution of several strategies to isoniazid (Manjunatha, Boshoff and Barry 2009). The efficacy of
detect PG or PG fragments (Humann and Lenz 2009; Otten these drugs highlights the essentiality of the mycobacterial cell
et al. 2018). The germline-encoded pattern-recognition recep- wall PG and highlights the opportunity to develop novel chem-
tors (PRRs) are expressed mainly on immune cells and recognise ical entities, which target enzymes involved in the synthesis of
pathogen-associated molecular patterns (PAMPs) and mediate the mycobacterial cell envelope (Feng and Barletta 2003; Silver
the production of immunoregulatory cytokines such as tumour 2006; Prosser and de Carvalho 2013). Further research to iden-
necrosis factor (TNF) and type I interferons (IFNs) (Medzhi- tify the unique features of the mycobacterial PG enzymes with
tov and Janeway 1997). Toll-like receptors (TLR) are activated respect to their structure, function and regulation can advance
by immunomodulatory lipoproteins in mycobacteria such as the development of narrow-spectrum, targeted antibiotics.
lipomannan (LM), lipoarabinomannan (LAM), mannosyl capped To date the early stages of PG biosynthesis have been poorly
LAM (ManLAM) or phosphoinositide capped LAM (PILAM) and PG exploited as antibacterial targets, and so present a significant
components present on the cell surface or endosome/lysosome opportunity for the development of the next generation of
membranes (Gilleron, Quesniaux and Puzo 2003; Quesniaux et al. antimycobacterial drugs. In particular, the enzymes involved in
2004; Singh et al. 2012; Esin et al. 2013; Xie et al. 2013). The cyto- the cytoplasmic stage of the PG biosynthetic pathway are cur-
plasm of the cell is also under scrutiny for PAMPs that breach the rently considered as new targets for drug discovery. This inter-
cell membrane. These PAMPs are recognised by constitutively est, however, has yet to translate to the development of a clin-
expressed Nod-like receptors (NLR) like NOD, NALP and NAIP. ically useful molecule. Although inhibitors against MurA and
They consist of the NACHT or NOD oligomerisation domain and MurB have been reported previously (Baum et al. 2001; Bronson
a motif similar to the microbial sensing leucine-rich repeat (LRR) et al. 2003; Yang et al. 2006) (1, 2, 3, Fig. 6.) herein, we will review
receptors present in the TLRs. While NOD1 specifically recog- the progress made to date towards the development of inhibitors
nises the γ -d-glutamyl-mDAP peptide of PG fragments, NOD2 targeting the ATP-dependent Mur ligases MurC–F involved in the
and NALP3 are activated by intracellular muramyl dipeptides cytoplasmic phase of PG biosynthesis.
(Mukamolova et al. 1998a; Zhang et al. 2001; Chamaillard et al. The Mur ligases are found in all clinically significant bacte-
2003a; Girardin et al. 2003a; Chamaillard et al. 2003b; Uehara et al. rial pathogens, and so inhibitors would be expected to be broad
2006; Humann and Lenz 2009; Pandey et al. 2009; Killick et al. spectrum. In addition, (i) there are conserved structural and
2013; Machowski et al. 2014). Thus, any changes in the structure sequence motifs and 3 binding sites in MurC-F against which
of PG could impair the innate immune responses of the host. antimicrobial agents can be designed (e.g. MurE Fig. 3) and (ii)
partial inhibition at multiple steps of PG synthesis (various Mur
enzymes) compared to inhibition of a single enzyme in the path-
Targeting PG biosynthesis to tackle antibiotic resistance
way may provide a more efficient route to obtaining bacterici-
(R) dal activity and decrease the likelihood of development of resis-
The ‘Golden Age’ of antibacterial drug discovery appears to be
over: since the 1990s there has been a void in the development tance. As a result, pan or multi-target inhibitors are conceivable
of new drugs for the treatment of bacterial infections (Silver and these may provide a better approach to obtaining robust
2011). More specifically, for TB the current frontline treatment antibacterial activity.
regimen is a cocktail of four drugs (isoniazid, rifampicin, pyraz- Adding to the appeal of the ATP-dependent Mur ligases as
inamide and ethambutol) that was first introduced in the 1960’s targets for drug design, many X-ray crystal structures have been
(Zumla, Nahid and Cole 2013). The emergence of MDR- and XDR- solved for proteins from different bacterial species (Bertrand
TB strains, however, have reinvigorated the search for antimy- et al. 1999; Deva et al. 2006; Basavannacharya et al. 2010a). How-
cobacterial agents with novel modes of action (Young et al. 2008). ever, only the X-ray crystal structure of MurE from M. tubercu-
Recent progress in this area includes the approval of Bedaquiline losis is currently available (Basavannacharya et al. 2010a; Basa-
(Sirturo) (Mahajan 2013; Koul et al. 2014) and Delamanid (Del- vannacharya et al. 2010b). Homology models of MurC and MurD
tyba) (Ryan and Lo 2014). from M. tuberculosis and MurC-F from M. leprae have been built
Pathways involved in the biosynthesis of cell wall PG have based on sequence identification with the corresponding homo-
historically been recognised as important drug targets. This is logue from either E. coli or H. influenzae (Anuradha et al. 2010;
demonstrated by the fact that a significant number of the antibi- Arvind et al. 2012; Shanmugam, Anbazhagan and Natarajan
otics in current clinical use, primarily β-lactams (e.g. penicillins 2012); Shanmugam and Natarajan 2012). Docking of native sub-
and cephalosporins) and glycopeptides, act by inhibiting the strates to these models aligned well with known X-ray crystal
later stages of PG biosynthesis (Fig. 2). Mycobaterium tuberculosis structures and good homology was observed between conserved
is considered resistant to many β-lactams, due to a combina- binding site residues (Anuradha et al. 2010; Arvind et al. 2012;
tion of the following factors: the thick, complex and hydropho- Shanmugam and Natarajan 2012). In silico docking of naphtha-
bic nature of its cell envelope, the presence of one or more lene sulphonamide inhibitors recapitulated the binding mode
active β-lactamases (Nampoothiri et al. 2008) and low-affinity observed in X-ray crystal structures and highlighted the impor-
PBPs (Chambers et al. 1995). A number of clinically useful TB tance of the Glu moiety as a driver of affinity for MurD (Arvind
drugs, however, do target cell wall processes. These include: (i) et al. 2012). One may expect that inhibitors of Mur ligases
the first line anti-TB drug isoniazid which inhibits MA biosyn- from other species may also inhibit the mycobacterial enzymes.
thesis (Zhang et al. 1992), (ii) ethambutol, another frontline drug, These models, therefore, provide a good starting point for the
which inhibits AG synthesis (Mikusova et al. 1995) and (iii) a sec- design of novel ligands, however, experimental validation of the
ond line drug d-cycloserine which is a broad spectrum antibi- predicted structures is currently lacking.
otic that inhibits PG biosynthesis via inhibition of Alr and Ddl The progress made to date in the design of Mur ligase
(Zhang 2005). Delamanid, like isoniazid, also targets genes in MA inhibitors has recently been reviewed, although it is notewor-
biosynthesis (Ryan and Lo 2014); and Pretomanid (PA-824), an thy that activity data on M. tuberculosis are woefully missing (Sil-
antitubercular drug under regulatory review by the FDA, has also ver 2011; Perdih et al. 2014; Sangshetti et al. 2017). The inhibitors
been shown to affect not only respiratory genes, but also tran- discovered to date fall primarily into three classes: (i) transi-
scription of genes responsive to known cell wall inhibitors like tion state analogues, which contain for example phosphinate
Maitra et al. 563

Figure 6. Structures of representative inhibitors of MurA-F with inhibitory data.

or sulphonamide functional groups which mimic the tetrahe- 2012) and (iii) inhibitors that bind to the ATP binding site
dral transition state formed during the enzyme-catalysed reac- (Hameed P et al. 2014). A number of core functional groups are
tion (Fig. 3) (Marmor et al. 2001; Reck et al. 2001), (ii) phos- repeatedly found in inhibitors of MurA to F, particularly those
phate/diphosphate mimics which typically contain a hetero- which act as mimics of the tetrahedral transition state or are
cyclic moiety (e.g. rhodanine, thiazolidindione) (Barreteau et al. phosphate bioisosteres. The majority of inhibitors developed
564 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

to date have arisen from HTS ( in vitro or in silico) followed by growth, further investigation, however suggested a pleiotropic
structure-guided design, however, natural products have also mechanism of action (Pesnot et al. 2011; Guzman et al. 2015).
been the source of a number of inhibitors (Guzman et al. 2010; A significant number of potent nanomolar inhibitors have
Guzman et al. 2013b). been developed for MurF. Abbott Laboratories, for example,
The majority of published MurC inhibitors are either phos- developed a series of cyanothiophene ligands with micromolar
phinate transition state analogues (4, Fig. 6) (Marmor et al. 2001; to nanomolar affinity (10, Fig. 6) (Gu et al. 2004; Stamper et al.
Reck et al. 2001) or l-Ala analogues (El Zoeiby et al. 2003). To the 2006) and Johnson & Johnson developed a series of potent thi-
best of our knowledge, however, antibacterial activity has not azolylaminopyrimidine containing inhibitors (11, Fig. 6) (Baum
been demonstrated for either class, which is perhaps unsurpris- et al. 2006). However, none of the compounds tested demon-
ing as the above compounds are very polar and are unlikely to strated any significant antibacterial activity. Finally, the identifi-
be membrane permeable. The most promising MurC inhibitors cation of diarylquinolines as low micromolar inhibitors of MurF
identified to date were reported in a study by Astra Zeneca in led to the development of 12 (Fig. 6), which exhibited an MIC
which a series of potent pyrazolopyrimidines were developed of 8 μg/mL against a number of bacterial species (Enterococcus
(Fig. 6, 5). Compound 5 was proposed to bind into the ATP bind- faecalis, E. faecium and S. aureus). Compound 12 binds MurF in
ing site and encouragingly, also demonstrated bactericidal activ- the presence of ATP and so is likely to occupy the substrate
ity, albeit against efflux deficient mutants (MIC = 3.1 and 1.5 binding site. In addition, treatment of LPS deficient E. coli with
μM against E. coli and P. aeruginosa respectively). Importantly, 5 12 resulted in the accumulation of the substrate of MurF, how-
also demonstrated selectivity by not inhibiting a panel of human ever, over expression of MurF did not significantly affect the
kinases. From mutation resistance maps 5 was suggested to be MIC. Hence, although the diarlyquinolines represent a promis-
selective for MurC over the other Mur ligases, however, biochem- ing start point for the development of novel antibacterial agents,
ical data to test for inhibition of MurD-F is lacking. Most signifi- the mechanism of action warrants further examination.
cantly, the mode of action of the pyrazolopyrimidines was con- Strategies for the development of multi-target inhibitors of
clusively demonstrated to be as a result of inhibition of MurC. the Mur ligases fall in to two categories: (i) ligands that mimic
This is a particularly noteworthy advance, as it is the first exam- the shared tetrahedral intermediate of the Mur ligases and
ple in which an antibacterial activity of a Mur ligase inhibitor has (ii) ligands designed to target the ATP-binding site, which has
been demonstrated to be due to target engagement (Hameed P the greatest sequence and structural homology amongst the
et al. 2014). enzymes. Alternatively, for dual target inhibitors, the substrate
MurC phosphinate transition state analogues are also potent, binding sites of MurC/D and MurE/F are likely to have sufficient
sub-nanomolar inhibitors of MurD. Replacement of the uridine similarity to allow the design of dual inhibitors. The majority
core with an aryl sulphonamide as a phosphate mimic afforded of efforts to date have focussed on the first strategy leading to
the simplified inhibitor 6 (Fig. 6), which retained an IC50 in the benzene-1,3-dicarboxylic acid 2,5-dimethylpyrrole derivatives
low micromolar range against MurD and was more amenable to (Fig. 6, 13 & 14) (Perdih et al. 2014) and napthyl tetronic acids
optimisation by medicinal chemistry (Strancar et al. 2006). The d- (Mansour et al. 2007) as first multi-target lead compounds. To
glutamate moiety or conformationally restrained analogues are evaluate the potential of these Mur ligase inhibitors as inhibitors
present in the majority of published inhibitors, as they are key of the mycobacterial enzymes, we performed a docking study
to potent inhibition of MurD (7, 8, Fig. 6.) (Barreteau et al. 2012). (Autodock Vina) (Trott and Olson 2010), using a select number
Another class of MurD inhibitors feature a benzylidene rhoda- of inhibitors and MurE (pdb code: 2XJA) from M. tuberculosis as a
nine moiety, which occupies the uridine diphosphate binding model system. The pyrazalopyrimidine 5 was designed to inhibit
site (7, Fig. 6). Although 7 is a low micromolar inhibitor of the E. MurC from E. coli (Deva et al. 2006). Docking of 5 to MurE from
coli enzyme, it inhibits the mycobacterial enzyme with an IC50 of M. tuberculosis resulted in the prediction of two major binding
200 μM, highlighting the challenges associated with the design modes. In the first binding mode there is good overlap between
of broad spectrum agents (Barreteau et al. 2012). the pyrazolpyrimidine ring and the adenosine ring of ATP (as
A number of the reported MurD inhibitors also inhibit MurE observed X-ray crystal structure) allowing a π-π stacking inter-
and, again, contain phosphinate groups (or sulphonamides) action with Tyr343 (an aromatic residue at this position is highly
that mimic the tetrahedral intermediate formed during the conserved amongst the Mur ligases); the pyrazole ring is pre-
enzyme-catalysed reaction (Tanner et al. 1996; Strancar et al. dicted to occupy the phosphate binding site and forms a H-
2007). Alternative scaffolds have been identified from natu- bonding interaction with Thr158 (part of the conserved diphos-
ral product sources and are of particular interest here due to phate binding motif) (Fig. 7). In the second binding mode the
their demonstrated antimycobacterial activity. For example N- pyrazole ring forms a π-π stacking interaction with Tyr343, the
methyl-2-alkenyl-4-quinolone derivatives isolated from Evodia pyrazolopyrimidine moiety is predicted to form polar contacts
rutaecarpaand aporphine alkaloids isolated from Ocotea macro- with two key residues (Asn347 and Thr158) and the hydroxyl
phylla inhibit the activity of MurE from M. tuberculosis (9, Fig. 6,) group interacts with residues in the highly conserved phosphate
and prevent cell growth at micromolar concentrations (Guz- binding loop (Gly156, Lys157, Thr158) (Fig. 7B). Given the reca-
man et al. 2010; Guzman et al. 2011; Wube et al. 2011; Guzman pitulation of key interactions formed by ATP within the bind-
et al. 2013b). These natural products also inhibit the growth of ing site and similarities with the binding mode predicted by
a number of other Gram-positive and Gram-negative bacteria, Hameed et al. it is feasible that 5 and analogues thereof may pro-
but at much higher concentrations than those required against vide a starting points for the development of inhibitors for the
mycobacteria. It has yet to be demonstrated that the observed mycobacterial Mur ligases (Hameed P et al. 2014).
antibacterial activity is due to inhibition of MurE only, however, The benzylidene rhodanine inhibitor 7 was designed as an
given the relatively low affinity and lipophilicity of these com- inhibitor of MurD from E. coli (Zidar et al. 2010). In the X-ray
pounds it is unlikely that they are highly selective for their tar- crystal structure 7 was found to span the substrate binding site
get. The natural product S-leucoxine from Rhodostemonodaphne with the rhodanine heterocycle acting as a diphosphate mimic.
crenaticupula and related synthetic 1-tetrahydroquinolines have This binding mode is recapitulated when docked into MurE from
also been demonstrated to inhibit MurE and mycobacterial cell M. tuberculosis, however, because of the larger binding site the
Maitra et al. 565

Figure 7. Representative predictive binding pockets for selected compounds to MurE (PDB code 2XJA) from M. tuberculosis (overlaid with ATP or UAG as appropriate,
shown in lines) (A), Compound 5, predicted binding affinity −9.2 kcal/mol (B), Compound 5, predicted binding affinity −8.9 kcal/mol (C), Compound 8, predicted binding
affinity -7.8 kcal/mol (D) Compound 8, predicted binding affinity −7.2 kcal/mol (E) Compound 14, predicted binding affinity −6.8 kcal/mol.

rhodanine heterocycle is predicted to bind in the hydrophobic well onto the observed conformation of UDP-MurNac-peptide
pocket occupied by the uracil moiety of UAG and not to the in the X-ray crystal structure of MurE (Fig. 7C, D). However, it
diphosphate binding site as in E. coli MurD. is unlikely that the sulphonamide moiety will be able to bind
Transition state analogue, 8, another inhibitor of MurD, in a similar conformation to the tetrahedral transition state due
which contains a conformationally restrained Glu analogue and to the larger and more open binding site of MurE in compari-
a sulphonamide moiety as a transition state analogue, was also sion to MurD. It is important to note that the N-terminal domain
docked to MurE. This inhibitor was predicted to span the sub- exhibits the greatest sequence and structural variation i.e. the
strate binding site and two alternate binding modes were pre- N-terminal domain in MurC/D is significantly different to that in
dicted: (i) in which the Glu mimic occupied the uracil binding MurE/F. This domain is also more flexible and undergoes signifi-
site and the sulphonamide the diphosphate binding site and (ii) cant conformational changes on ligand binding; this flexibility is
in which the cyanobenzyl functional group occupied the uracil required to enable the Mur ligase to accommodate the growing
binding site and the sulphonamide and Glu mimic mapped peptide chain of the PG precursor (Basavannacharya et al. 2010b).
566 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

As docking experiments are based solely on and restricted by the Fragment-based drug discovery may offer a viable alternative
structures/conformations available, these need to be integrated (Hajduk and Greer 2007). This approach employs libraries of low
with in vitro experiments such as enzyme inhibition kinetics to molecular weight compounds or fragments, these could include
gain confidence in the targeting of inhibitors. commercial fragment libraries, repurposed kinase focussed
A related set of compounds designed as multi-target Mur lig- libraries to target the ATP binding site and libraries of phos-
ase inhibitors are compounds 13 (a dual inhibitor of MurD/E) phate bioisosteres (Huang et al. 2009; Volkamer et al. 2015). The
and 14 (an inhibitor of MurC-F). These were docked against MurE fragments are screened against the protein targets of interest
from M. tuberculosis (Tomasic et al. 2010; Basavannacharya et al. using a range of biophysical assays which have a much lower
2010a; Perdih et al. 2014). For 13, as expected, the dihydopyrim- false positive hit rate compared to the biochemical assays typ-
idine moiety was predicted to act as a diphosphate mimic and ically used in HTS screens (Schulz and Hubbard 2009). One of
the phenyl ring to occupy the uracil binding site. The remain- the major advantages of this approach, and which is partic-
der of the inhibitor maps well onto the conformation of UAG ularly relevant to antimicrobial drug discovery, is that due to
observed in the X-ray crystal structure ( 13 is, however, trun- the nature of the hits identified, i.e. low molecular weight, low-
cated compared to UAG and so does not span the entire binding affinity and high-ligand efficiency, one can exert greater con-
site). 14, which is a moderate inhibitor of MurC-F, is predicted to trol over the physiochemical properties of the inhibitor dur-
adopt a similar conformation within the substrate binding site ing the process of hit to lead development (Hajduk 2006). The
with the rhodanine heterocycle again acting as a diphosphate process of fragment optimisation is typically design intensive
mimic and the chlorophenyl moiety occupying the uracil bind- and structure guided, however, given the current lack of struc-
ing site. The increased flexibility in 14, compared to its precur- tural information for the mycobacterial enzymes, in situ opti-
sor 13, allows the heterocycle to form polar contacts with key misation using kinetic target guided synthesis has great poten-
residues in the diphosphate binding loop (Gly83, Ser84). Interac- tial and would allow rapid exploration of the protein bind-
tions with key residues in the substrate binding site are picked ing landscape and the inherent flexibility of the Mur ligases
up by the isophthalic acid moiety (Arg230, Ser222, Thr195 and (Hajduk 2006).
Thr196) (Fig. 7E). The recapitulation of key interactions between It is clear from the progress to date that to obtain anti-
substrate and protein supports the conclusion that these lig- tubercular agents that target the Mur ligases a number of hur-
ands likely bind to and inhibit mycobacterial MurE, although the dles need to be overcome. As a result, further exploration of
inherent flexibility of the Mur ligases is likely to complicate this the Mur ligases as drug targets and the mechanisms of the
picture. Experimental evidence is needed to confirm inhibition inhibitors developed to date are necessary if therapeutically use-
of mycobacterial Mur ligases by any of the inhibitors discussed ful molecules are to be developed.
here.
As can be seen from this survey of the literature, much
progress towards the development of potent inhibitors of the
Mur ligases has been made. However, the translation from effec-
CONCLUSIONS AND FUTURE OUTLOOK
tive enzyme inhibition to antibacterial action is a significant Due to the protective role of PG and its absence in humans,
challenge. Membrane permeability and active efflux are likely its biosynthetic pathway remains an excellent therapeutic tar-
to present the biggest hurdles and many inhibitors developed to get (Strancar et al. 2007; Zawadzke et al. 2008; Tomasic et al.
date are very polar, particularly the phosphinate transition state 2010). Encouragingly, technologies for high throughput screen-
analogues and amino acid derivatives, making them unlikely ing against these targets have advanced (Kristan et al. 2009; Turk
to easily traverse the membrane (Jarlier and Nikaido 1994). For et al. 2009; Guzman et al. 2013a). We would hope that these devel-
other classes of inhibitors, the explanation is less clear. opments and the new insights into the molecular mechanisms
It is also important to highlight that for those compounds of cell wall synthesis could lead to the development of targeted
with a demonstrated antibacterial activity there is often no evi- anti-mycobacterials.
dence for the observed bactericidal effect being caused by inhibi-
tion of a Mur ligase (Silver 2011). The IC50 /MIC data in conjunc-
tion with molecular modelling or X-ray crystallography, would
significantly inform the design of next generation of inhibitors. SUPPLEMENTARY DATA
The study by Barreteau et al. (Barreteau et al. 2012), which eval- Supplementary data are available at FEMSRE online
uated a selection of inhibitors designed to target MurD from E.
coli against orthologues from different bacteria is a step in this
direction.
Many of the inhibitors discussed contain what would typ- ACKNOWLEDGEMENTS
ically be considered undesirable functional groups, for exam-
We are grateful to Professor Simon Gibbons and Professor Tim-
ple rhodanine and thiazolidindione moieties are notorious
othy D. McHugh for their helpful suggestions. SB would like
pan assay interference compounds (PAINS) (Baell 2010; Baell
to thank MRC, UK for a New Investigators Research Grant
and Holloway 2010). These compounds often prove to be not
(GO801956). AM and LTM would like to thank the Wellcome Trust
amenable to optimisation, preventing the progression of hit/lead
(108877/Z/15/Z) for their PhD studentship. WV received support
molecules. The pitiful success of HTS as a source of novel
from the BBSRC (BB/P001289/1).
antibacterial agents has been the subject of a comprehen-
The authors acknowledge that the work of many eminent
sive commentary in the literature (Tomašić and Mašič 2012).
researchers could not be cited in the review due to limited
Hits derived from HTS campaigns are typically quite lipophilic,
space. We are grateful for their collective research contribut-
whereas antibiotics are typically larger and more polar than
ing to advances in technology and our understanding of the
drugs designed for human targets (Payne et al. 2007). It is, there-
mycobacterial cell envelope.
fore, questionable whether the libraries amassed by the phar-
maceutical industry are optimal for antibacterial drug discovery. Conflict of interest. None declared.
Maitra et al. 567

REFERENCES Becker K, Sander P. Mycobacterium tuberculosis lipoproteins in vir-


ulence and immunity–fighting with a double-edged sword.
Ahangar MS, Furze CM, Guy CS et al. Structural and functional
FEBS Lett 2016;590:3800–19.
determination of homologs of the Mycobacterium tuberculosis
Benson TE, Marquardt JL, Marquardt AC et al. Overexpres-
N-acetylglucosamine-6-phosphate deacetylase (NagA). J Biol
sion, purification, and mechanistic study of UDP-N-
Chem 2018;293:9770–83.
acetylenolpyruvylglucosamine reductase. Biochemistry
Al-Dabbagh B, Henry X, Ghachi ME et al. Active Site Map-
1993;32:2024–30.
ping of MraY, a Member of the Polyprenyl-phosphate N-
Bergfeld AK, Pearce OM, Diaz SL et al. Metabolism of ver-
Acetylhexosamine 1-Phosphate Transferase Superfamily,
tebrate amino sugars with N-glycolyl groups: elucidating
Catalyzing the First Membrane Step of Peptidoglycan Biosyn-
the intracellular fate of the non-human sialic acid N-
thesis†. Biochemistry 2008;47:8919–28.
glycolylneuraminic acid. J Biol Chem 2012;287:28865–81.
Aldridge BB, Fernandez-Suarez M, Heller D et al. Asymmetry
Bertrand JA, Auger G, Martin L et al. Determination of the MurD
and aging of mycobacterial cells lead to variable growth and
mechanism through crystallographic analysis of enzyme
antibiotic susceptibility. Science 2012;335:100–4.
complexes. J Mol Biol 1999;289:579–90.
Anderson MS, Eveland SS, Onishi HR et al. Kinetic mecha-
Bhakta S, Basu J. Overexpression, purification and biochemical
nism of the Escherichia coli UDPMurNAc-tripeptide D-alanyl-
characterization of a class A high-molecular-mass penicillin-
D-alanine-adding enzyme: use of a glutathione S-transferase
binding protein (PBP), PBP1∗ and its soluble derivative from
fusion. Biochemistry 1996;35:16264–9.
Mycobacterium tuberculosis. Biochem J 2002;361:635–9.
Anuradha CM, Mulakayala C, Babajan B et al. Probing ligand
Bhattacharjee N, Triboulet S, Dubée V et al. Negative impact
binding modes of Mycobacterium tuberculosis MurC ligase by
of carbapenem methylation on the reactivity of β-lactams
molecular modeling, dynamics simulation and docking. J Mol
for cysteine acylation revealed by quantum calculations and
Model 2010;16:77–85.
kinetic analyses. Antimicrob Agents Chemother 2019; AAC.
Arvind A, Kumar V, Saravanan P et al. Homology modeling,
e02039-18.
molecular dynamics and inhibitor binding study on MurD
Bickford JS, Nick HS. Conservation of the PTEN catalytic motif
ligase of Mycobacterium tuberculosis. Interdiscip Sci, Comput Life
in the bacterial undecaprenyl pyrophosphate phosphatase,
Sci 2012;4:223–38.
BacA/UppP. Microbiology 2013;159:2444–55.
Awasthy D, Bharath S, Subbulakshmi V et al. Alanine racemase
Bielnicki J, Devedjiev Y, Derewenda U et al. B. subtilis ykuD pro-
mutants of Mycobacterium tuberculosis require D-alanine for
tein at 2.0 A resolution: insights into the structure and func-
growth and are defective for survival in macrophages and
tion of a novel, ubiquitous family of bacterial enzymes. Pro-
mice. Microbiology 2012;158:319–27.
teins 2006;62:144–51.
Babajan B, Chaitanya M, Rajsekhar C et al. Comprehensive struc-
Bolla JR, Sauer JB, Wu D et al. Direct observation of the influ-
tural and functional characterization of Mycobacterium tuber-
ence of cardiolipin and antibiotics on lipid II binding to MurJ.
culosis UDP-NAG enolpyruvyl transferase (Mtb-MurA) and
Nature Chem 2018;10:363.
prediction of its accurate binding affinities with inhibitors.
Botella H, Vaubourgeix J, Lee MH et al. Mycobacterium tuberculo-
Interdiscip Sci, Comput Life Sci 2011;3:204–16.
sis protease MarP activates a peptidoglycan hydrolase during
Baell JB, Holloway GA. New substructure filters for removal of
acid stress. EMBO J 2017a;36:536–48.
pan assay interference compounds (PAINS) from screening
Botella H, Yang G, Ouerfelli O et al. Distinct spatiotemporal
libraries and for their exclusion in bioassays. J Med Chem
dynamics of peptidoglycan synthesis between Mycobac-
2010;53:2719–40.
terium smegmatis and Mycobacterium tuberculosis. MBio
Baell JB. Observations on screening-based research and some
2017b;8:e01183–17.
concerning trends in the literature. Future Med Chem
Both D, Schneider G, Schnell R. Peptidoglycan remodeling in
2010;2:1529–46.
Mycobacterium tuberculosis: comparison of structures and cat-
Baranowski C, Welsh MA, Sham L-T et al. Maturing Mycobacterium
alytic activities of RipA and RipB. J Mol Biol 2011;413:247–60.
smegmatis peptidoglycan requires non-canonical crosslinks
Both D, Steiner EM, Stadler D et al. Structure of LdtMt2, an L,D-
to maintain shape. eLife 2018;7:e37516.
transpeptidase from Mycobacterium tuberculosis. Acta Crystal-
Barreteau H, Sosic I, Turk S et al. MurD enzymes from dif-
logr Sec D, Biol crystallogr 2013;69: 432–41.
ferent bacteria: evaluation of inhibitors. Biochem Pharmacol
Boudreau MA, Fisher JF, Mobashery S. Messenger functions of
2012;84:625–32.
the bacterial cell wall-derived muropeptides. Biochemistry
Basavannacharya C, Moody PR, Munshi T et al. Essential
2012;51:2974–90.
residues for the enzyme activity of ATP-dependent MurE
Bourai N, Jacobs WR, Jr., Narayanan S. Deletion and overex-
ligase from Mycobacterium tuberculosis. Protein Cell 2010b;1:
pression studies on DacB2, a putative low molecular mass
1011–22.
penicillin binding protein from Mycobacterium tuberculosis
Basavannacharya C, Robertson G, Munshi T et al. ATP-dependent
H(37)Rv. Microb Pathog 2012;52: 109–16.
MurE ligase in Mycobacterium tuberculosis: biochemical and
Boutte CC, Baer CE, Papavinasasundaram K et al. A cytoplasmic
structural characterisation. Tuberculosis (Edinb) 2010a;90:16–
peptidoglycan amidase homologue controls mycobacterial
24.
cell wall synthesis. eLife 2016;5:e14590.
Baum EZ, Crespo-Carbone SM, Abbanat D et al. Utility of
Brennan PJ, Nikaido H. The envelope of mycobacteria. Annu Rev
muropeptide ligase for identification of inhibitors of the cell
Biochem 1995;64:29–63.
wall biosynthesis enzyme MurF. Antimicrob Agents Chemother
Brennan PJ. Structure, function, and biogenesis of the cell wall of
2006;50:230–6.
Mycobacterium tuberculosis. Tuberculosis (Edinb) 2003;83:91–7.
Baum EZ, Montenegro DA, Licata Let al. Identification and
Bronson JJ, DenBleyker KL, Falk PJet al. Discovery of the first
characterization of new inhibitors of the Escherichia coli
antibacterial small molecule inhibitors of MurB. Bioorg. Med.
MurA enzyme. Antimicrob. Agents Chemother 2001; 45:3182–
Chem. Lett. 2003;13 :873–5. 12617911
8. 11600375
568 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

Brown ED, Vivas EI, Walsh CT et al. MurA (MurZ), the enzyme that Crick DC, Schulbach MC, Zink EE et al. Polyprenyl phosphate
catalyzes the first committed step in peptidoglycan biosyn- biosynthesis in Mycobacterium tuberculosis and Mycobacterium
thesis, is essential in Escherichia coli. J Bacteriol 1995;177: 4194– smegmatis. J Bacteriol 2000;182:5771–8.
7. Dajkovic A, Tesson B, Chauhan S et al. Hydrolysis of peptidogly-
Bruning JB, Murillo AC, Chacon O et al. Structure of the Mycobac- can is modulated by amidation of meso-diaminopimelic acid
terium tuberculosis D-alanine:D-alanine ligase, a target of and Mg(2+) in Bacillus subtilis. Mol Microbiol 2017;104:972–88.
the antituberculosis drug D-cycloserine. Antimicrob Agents Daniel RA, Errington J. Control of cell morphogenesis in bac-
Chemother 2011;55:291–301. teria: two distinct ways to make a rod-shaped cell. Cell
Carel C, Nukdee K, Cantaloube S et al. Mycobacterium tuberculo- 2003;113:767–76.
sis proteins involved in mycolic acid synthesis and transport Darby CM, Venugopal A, Ehrt S et al. Mycobacterium tuberculosis
localize dynamically to the old growing pole and septum. gene Rv2136c is dispensable for acid resistance and virulence
PLoS One 2014;9:e97148. in mice. Tuberculosis 2011;91:343–7.
Cava F, de Pedro MA, Lam H et al. Distinct pathways for modi- Dasgupta A, Datta P, Kundu M et al. The serine/threonine kinase
fication of the bacterial cell wall by non-canonical D-amino PknB of Mycobacterium tuberculosis phosphorylates PBPA, a
acids. EMBO J 2011;30:3442–53. penicillin-binding protein required for cell division. Microbi-
Chalker AF, Ingraham KA, Lunsford RD et al. The bacA gene, ology 2006;152:493–504.
which determines bacitracin susceptibility in Streptococcus David S. Synergic activity of d-cycloserine and β-chloro-
pneumoniae and Staphylococcus aureus, is also required for vir- d-alanine against Mycobacterium tuberculosis. J Antimicrob
ulence. Microbiology 2000;146:1547–53. Chemother 2001;47:203–6.
Chaloupka J, Strnadova M. Turnover of murein in a DeJesus MA, Gerrick ER, Xu W et al. Comprehensive essentiality
diaminopimelic acid dependent mutant of Escherichia analysis of the Mycobacterium tuberculosis genome via satu-
coli. Folia Microbiol (Praha) 1972;17:446–55. rating transposon mutagenesis. MBio 2017;8:e02133–16.
Chamaillard M, Girardin SE, Viala J et al. Nods, Nalps and Naip: Dementin S. Etude du mécanisme réactionnel des Mur
intracellular regulators of bacterial-induced inflammation. synthétases, enzymes impliquées dans la biosynthèse
Cell Microbiol 2003a;5:581–92. du peptidoglycane bactérien. 2001.
Chamaillard M, Hashimoto M, Horie Y et al. An essential role for den Blaauwen T, de Pedro MA, Nguyen-Disteche M et al.
NOD1 in host recognition of bacterial peptidoglycan contain- Morphogenesis of rod-shaped sacculi. FEMS Microbiol Rev
ing diaminopimelic acid. Nat Immunol 2003b;4:702–7. 2008;32:321–44.
Chambers HF, Moreau D, Yajko D et al. Can penicillins and other De Smet KA, Kempsell KE, Gallagher A et al. Alteration of a
beta-lactam antibiotics be used to treat tuberculosis? Antimi- single amino acid residue reverses fosfomycin resistance of
crob Agents Chemother 1995;39:2620–4. recombinant MurA from Mycobacterium tuberculosis. Microbi-
Chao MC, Kieser KJ, Minami S et al. Protein complexes and prote- ology 1999;145:3177–84.
olytic activation of the cell wall hydrolase RipA regulate sep- Deva T, Baker EN, Squire CJ et al. Structure of Escherichia coli
tal resolution in mycobacteria. PLoS Pathog 2013;9:e1003197. UDP-N-acetylmuramoyl:L-alanine ligase (MurC). Acta crystal-
Chauviac FX, Robertson G, Quay DH et al. The RpfC (Rv1884) lographica Section D, Biological crystallography 2006;62:1466–74.
atomic structure shows high structural conservation within Dmitriev B, Toukach F, Ehlers S. Towards a comprehensive view
the resuscitation-promoting factor catalytic domain. Acta of the bacterial cell wall. Trends Microbiol 2005;13:569–74.
Crystallogr F Struct Biol Commun 2014;70:1022–6. Dmitriev BA, Toukach FV, Schaper KJ et al. Tertiary struc-
Cheng Q, Park JT. Substrate specificity of the AmpG permease ture of bacterial murein: the scaffold model. J Bacteriol
required for recycling of cell wall anhydro-muropeptides. J 2003;185:3458–68.
Bacteriol 2002;184:6434–6. Dominguez-Escobar J, Chastanet A, Crevenna AH et al. Proces-
Chesnokova ON, McPherson SA, Steichen CT et al. The spore- sive movement of MreB-associated cell wall biosynthetic
specific alanine racemase of Bacillus anthracis and its role in complexes in bacteria. Science 2011;333:225–8.
suppressing germination during spore development. J Bacte- Domı́nguez-Escobar J, Chastanet A, Crevenna AH et al. Proces-
riol 2009;191:1303–10. sive movement of MreB-associated cell wall biosynthetic
Chung BC, Zhao J, Gillespie RA et al. Crystal structure of MraY, an complexes in bacteria. Science 2011;333:225–8.
essential membrane enzyme for bacterial cell wall synthesis. Downing KJ, Mischenko VV, Shleeva MO et al. Mutants of
Science 2013;341:1012–6. Mycobacterium tuberculosis lacking three of the five rpf-like
Cohen-Gonsaud M, Barthe P, Bagneris C et al. The structure of a genes are defective for growth in vivo and for resuscitation
resuscitation-promoting factor domain from Mycobacterium in vitro. Infect Immun 2005;73:3038–43.
tuberculosis shows homology to lysozymes. Nat Struct Mol Biol Dubee V, Triboulet S, Mainardi JL et al. Inactivation of Mycobac-
2005;12:270–3. terium tuberculosis L,D-Transpeptidase Ldt(Mt1) by Car-
Cohen-Gonsaud M, Keep NH, Davies AP et al. Resuscitation- bapenems and Cephalosporins. Antimicrob Agents Chemother
promoting factors possess a lysozyme-like domain. Trends 2012;56:4189–95.
Biochem Sci 2004;29:7–10. Du W, Brown JR, Sylvester DR et al. Two active forms of UDP-N-
Cole ST, Brosch R, Parkhill J et al. Deciphering the biology acetylglucosamine enolpyruvyl transferase in gram-positive
of Mycobacterium tuberculosis from the complete genome bacteria. J Bacteriol 2000;182:4146–52.
sequence. Nature 1998;393:537–44. Dziadek B, Brzostek A, Grzybowski M et al. Mycobacterium tuber-
Craggs PD, Mouilleron S, Rejzek M et al. The mechanism of acetyl culosis AtsG (Rv0296c), GlmU (Rv1018c) and SahH (Rv3248c)
transfer catalyzed by Mycobacteri um tuberculosis GlmU. Bio- Proteins Function as the Human IL-8-Binding Effectors and
chemistry 2018;57:3387–401. Contribute to Pathogen Entry into Human Neutrophils. PLoS
Crick DC, Mahapatra S, Brennan PJ. Biosynthesis of the One 2016;11:e0148030.
arabinogalactan-peptidoglycan complex of Mycobacterium El Ghachi M, Derbise A, Bouhss A et al. Identification of multi-
tuberculosis. Glycobiology 2001;11:107R–18R. ple genes encoding membrane proteins with undecaprenyl
Maitra et al. 569

pyrophosphate phosphatase (UppP) activity in Escherichia coli. Gilleron M, Quesniaux VF, Puzo G. Acylation state of the
J Biol Chem 2005;280:18689–95. phosphatidylinositol hexamannosides from Mycobacterium
El Zoeiby A, Sanschagrin F, Darveau A et al. Identification of bovis bacillus Calmette Guerin and Mycobacterium tuberculosis
novel inhibitors of Pseudomonas aeruginosa MurC enzyme H37Rv and its implication in Toll-like receptor response. J Biol
derived from phage-displayed peptide libraries. J Antimicrob Chem 2003;278:29880–9.
Chemother 2003;51:531–43. Girardin SE, Boneca IG, Viala J et al. Nod2 is a general sensor of
Emami K, Guyet A, Kawai Y et al. RodA as the missing gly- peptidoglycan through muramyl dipeptide (MDP) detection.
cosyltransferase in Bacillus subtilis and antibiotic discov- J Biol Chem 2003a;278:8869–72.
ery for the peptidoglycan polymerase pathway. Nat Microbiol Girardin SE, Travassos LH, Herve M et al. Peptidoglycan molecu-
2017;2:16253. lar requirements allowing detection by Nod1 and Nod2. J Biol
Eniyan K, Dharavath S, Vijayan R et al. Crystal structure of UDP- Chem 2003b;278:41702–8.
N-acetylglucosamine-enolpyruvate reductase (MurB) from Glauner B, Holtje JV, Schwarz U. The composition of the murein
Mycobacterium tuberculosis. Biochimica et biophysica acta Proteins of Escherichia coli. J Biol Chem 1988;263:10088–95.
and proteomics 2018;1866:397–406. Goffin C, Ghuysen JM. Multimodular penicillin-binding proteins:
Eniyan K, Kumar A, Rayasam GV et al. Development of a one- an enigmatic family of orthologs and paralogs. Microbiol Mol
pot assay for screening and identification of Mur pathway Biol Rev 1998;62:1079–93.
inhibitors in Mycobacterium tuberculosis. Sci Rep 2016;6:35134. Goodell EW, Schwarz U. Release of cell wall peptides into culture
Erdemli SB, Gupta R, Bishai WR et al. Targeting the cell wall of medium by exponentially growing Escherichia coli. J Bacteriol
Mycobacterium tuberculosis: structure and mechanism of L,D- 1985;162:391–7.
transpeptidase 2. Structure 2012;20:2103–15. Goodell EW. Recycling of murein by Escherichia coli. J Bacteriol
Errington J. L-form bacteria, cell walls and the origins of life. Open 1985;163:305–10.
biology 2013;3:120143. Griffin JE, Gawronski JD, DeJesus MA et al. High-resolution
Esin S, Counoupas C, Aulicino A et al. Interaction of Mycobac- phenotypic profiling defines genes essential for mycobac-
terium tuberculosis cell wall components with the human nat- terial growth and cholesterol catabolism. PLoS Pathog
ural killer cell receptors NKp44 and Toll-like receptor 2. Scand 2011;7:e1002251.
J Immunol 2013;77:460–9. Grzegorzewicz AE, De Sousa-d’Auria C, McNeil MR et al. Assem-
Eveland SS, Pompliano DL, Anderson MS. Conditionally lethal bling of the Mycobacterium tuberculosis cell wall core. J Biol
Escherichia coli murein mutants contain point defects that Chem 2016;291:18867–79.
map to regions conserved among murein and folyl poly- Gupta R, Lavollay M, Mainardi JL et al. The Mycobacterium
gamma-glutamate ligases: identification of a ligase super- tuberculosis protein LdtMt2 is a nonclassical transpeptidase
family. Biochemistry 1997;36:6223–9. required for virulence and resistance to amoxicillin. Nat Med
Falk PJ, Ervin KM, Volk KS et al. Biochemical evidence for the 2010;16:466–9.
formation of a covalent acyl-phosphate linkage between Gu YG, Florjancic AS, Clark RF et al. Structure-activity relation-
UDP-N-acetylmuramate and ATP in the Escherichia coli UDP- ships of novel potent MurF inhibitors. Bioorganic & medicinal
N-acetylmuramate:L-alanine ligase-catalyzed reaction. Bio- chemistry letters 2004;14:267–70.
chemistry 1996;35:1417–22. Guzman JD, Evangelopoulos D, Gupta A et al. Antimycobacteri-
Feng Z, Barletta RG. Roles of Mycobacterium smegmatis D- als from lovage root (Ligusticum officinale Koch). Phytother-
alanine:D-alanine ligase and D-alanine racemase in the apy research: PTR 2013b;27:993–8.
mechanisms of action of and resistance to the peptido- Guzman JD, Evangelopoulos D, Gupta A et al. Antitubercular spe-
glycan inhibitor D-cycloserine. Antimicrob Agents Chemother cific activity of ibuprofen and the other 2-arylpropanoic acids
2003;47:283–91. using the HT-SPOTi whole-cell phenotypic assay. BMJ open
Figueiredo TA, Sobral RG, Ludovice AM et al. Identification of 2013a;3:e002672.
genetic determinants and enzymes involved with the amida- Guzman JD, Gupta A, Evangelopoulos D et al. Anti-tubercular
tion of glutamic acid residues in the peptidoglycan of Staphy- screening of natural products from Colombian plants:
lococcus aureus. PLoS Pathog 2012;8:e1002508. 3-methoxynordomesticine, an inhibitor of MurE lig-
Filippova EV, Kieser KJ, Luan CH et al. Crystal structures ase of Mycobacterium tuberculosis. J Antimicrob Chemother
of the transpeptidase domain of the Mycobacterium tuber- 2010;65:2101–7.
culosis penicillin-binding protein PonA1 reveal potential Guzman JD, Pesnot T, Barrera DA et al. Tetrahydroisoquinolines
mechanisms of antibiotic resistance. FEBS J 2016;283: affect the whole-cell phenotype of Mycobacterium tuberculo-
2206–18. sis by inhibiting the ATP-dependent MurE ligase. J Antimicrob
Flores AR, Parsons LM, Pavelka MS, Jr. Characterization of Chemother 2015;70:1691–703.
novel Mycobacterium tuberculosis and Mycobacterium smegma- Guzman JD, Wube A, Evangelopoulos D et al. Interaction of N-
tis mutants hypersusceptible to beta-lactam antibiotics. J methyl-2-alkenyl-4-quinolones with ATP-dependent MurE
Bacteriol 2005;187:1892–900. ligase of Mycobacterium tuberculosis: antibacterial activity,
Forrellad MA, Klepp LI, Gioffré A et al. Virulence factors of the molecular docking and inhibition kinetics. J Antimicrob
Mycobacterium tuberculosis complex. Virulence 2013;4:3–66. Chemother 2011;66:1766–72.
Garcia-Heredia A, Pohane AA, Melzer ES et al. Peptidogly- Hajduk PJ, Greer J. A decade of fragment-based drug design:
can precursor synthesis along the sidewall of pole-growing strategic advances and lessons learned. Nat Rev Drug Discov-
mycobacteria. eLife 2018;7:e37243. ery 2007;6:211.
Garner EC, Bernard R, Wang W et al. Coupled, circumferential Hajduk PJ. Fragment-based drug design: how big is too big? J Med
motions of the cell wall synthesis machinery and MreB fila- Chem 2006;49:6972–6.
ments in B. subtilis. Science 2011;333:222–5. Hameed P S, Manjrekar P, Chinnapattu M et al. Pyrazolopyrim-
Ghuysen JM. Serine beta-lactamases and penicillin-binding pro- idines establish MurC as a vulnerable target in Pseudomonas
teins. Annu Rev Microbiol 1991;45:37–67. aeruginosa and Escherichia coli. ACS Chem Biol 2014;9:2274–82.
570 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

Hansen JM, Golchin SA, Veyrier FJ et al. N-glycolylated pep- Inoue A, Murata Y, Takahashi H et al. Involvement of an essential
tidoglycan contributes to the immunogenicity but not gene, mviN, in murein synthesis in Escherichia coli. J Bacteriol
pathogenicity of Mycobacterium tuberculosis. J Infect Dis 2008;190:7298–301.
2014;209:1045–54. Jacobs C, Huang LJ, Bartowsky E et al. Bacterial cell wall recy-
Harth G, Masleša-Galić S, Tullius MV et al. All four Mycobac- cling provides cytosolic muropeptides as effectors for beta-
terium tuberculosis glnA genes encode glutamine synthetase lactamase induction. EMBO J 1994;13:4684–94.
activities but only GlnA1 is abundantly expressed and Jagtap PK, Soni V, Vithani N et al. Substrate-bound crystal
essential for bacterial homeostasis. Mol Microbiol 2005;58: structures reveal features unique to Mycobacterium tuber-
1157–72. culosis N-acetyl-glucosamine 1-phosphate uridyltransferase
Hayashi JM, Richardson K, Melzer ES et al. Stress-induced reor- and a catalytic mechanism for acetyl transfer. J Biol Chem
ganization of the mycobacterial membrane domain. MBio 2012;287:39524–37.
2018;9:e01823-17. Jankute M, Cox JA, Harrison J et al. Assembly of the mycobacterial
Heidrich C, Templin MF, Ursinus A et al. Involvement of N- cell wall. Annu Rev Microbiol 2015;69:405–23.
acetylmuramyl-L-alanine amidases in cell separation and Jarlier V, Nikaido H. Mycobacterial cell wall: structure and
antibiotic-induced autolysis of Escherichia coli. Mol Microbiol role in natural resistance to antibiotics. FEMS Microbiol Lett
2001;41:167–78. 1994;123:11–8.
Heidrich C, Ursinus A, Berger J et al. Effects of multiple deletions Johnson JW, Fisher JF, Mobashery S. Bacterial cell-wall recycling.
of murein hydrolases on viability, septum cleavage, and sen- Ann N Y Acad Sci 2013;1277:54–75.
sitivity to large toxic molecules in Escherichia coli. J Bacteriol Joyce G, Williams KJ, Robb M et al. Cell division site place-
2002;184:6093–9. ment and asymmetric growth in mycobacteria. PLoS One
Hernández-Rocamora VM, Otten CF, Radkov A et al. Coupling of 2012;7:e44582.
polymerase and carrier lipid phosphatase prevents product Kana BD, Gordhan BG, Downing KJ et al. The resuscitation-
inhibition in peptidoglycan synthesis. Cell Surface 2018;2:1– promoting factors of Mycobacterium tuberculosis are required
13. for virulence and resuscitation from dormancy but are
Herve M, Boniface A, Gobec S et al. Biochemical character- collectively dispensable for growth in vitro. Mol Microbiol
ization and physiological properties of Escherichia coli 2008;67:672–84.
UDP-N-acetylmuramate:L-alanyl-gamma-D-glutamyl- Kastrinsky DB, Barry CE. Synthesis of labeled meropenem for the
meso-diaminopimelate ligase. J Bacteriol 2007;189: 3987–95. analysis of M. tuberculosis transpeptidases. Tetrahedron Lett
Hett EC, Chao MC, Rubin EJ. Interaction and modulation of two 2010;51: 197–200.
antagonistic cell wall enzymes of mycobacteria. PLoS Pathog Kaur D, Brennan PJ, Crick DC. Decaprenyl diphosphate synthesis
2010;6:e1001020. in Mycobacterium tuberculosis. J Bacteriol 2004;186:7564–70.
Hett EC, Chao MC, Steyn AJ et al. A partner for the resuscitation- Kieser KJ, Boutte CC, Kester JC et al. Phosphorylation of the pep-
promoting factors of Mycobacterium tuberculosis. Mol Microbiol tidoglycan synthase PonA1 governs the rate of polar elonga-
2007;66:658–68. tion in mycobacteria. PLoS Pathog 2015;11:e1005010.
Hett EC, Rubin EJ. Bacterial growth and cell division: a mycobac- Kieser KJ, Rubin EJ. How sisters grow apart: mycobacterial
terial perspective. Microbiol Mol Biol Rev 2008;72:126–56, table growth and division. Nat Rev Microbiol 2014;12:550.
of contents. Kilburn JO, Best GK. Characterization of autolysins from
Ho H-T, Falk PJ, Ervin KM et al. UDP-N-acetylmuramyl-L-alanine Mycobacterium smegmatis. J Bacteriol 1977;129:750–5.
functions as an activator in the regulation of the Escherichia Killick KE, Ni Cheallaigh C, O’Farrelly C et al. Receptor-
coli glutamate racemase activity. Biochemistry 1995;34:2464– mediated recognition of mycobacterial pathogens. Cell Micro-
70. biol 2013;15:1484–95.
Holtje JV. Growth of the stress-bearing and shape-maintaining Kim HS, Kim J, Im HN et al. Structural basis for the inhi-
murein sacculus of Escherichia coli. Microbiol Mol Biol Rev bition of Mycobacterium tuberculosis L, D-transpeptidase by
1998;62:181–203. meropenem, a drug effective against extensively drug-
Huang D, Zhou T, Lafleur K et al. Kinase selectivity potential for resistant strains. Acta Crystallogr Sect D 2013;69:420–31.
inhibitors targeting the ATP binding site: a network analysis. Kluj RM, Ebner P, Adamek M et al. Recovery of the Peptidoglycan
Bioinformatics 2009;26:198–204. Turnover Product released by the Autolysin Atl in Staphylo-
Hugonnet J-E, Blanchard JS. Irreversible inhibition of the coccus aureus involves the Phosphotransferase System Trans-
Mycobacterium tuberculosis β-lactamase by clavulanate. Bio- porter MurP and the Novel 6-phospho-N-acetylmuramidase
chemistry 2007;46:11998–2004. MupG. Front Microbiol 2018;9.
Hugonnet J-E, Mengin-Lecreulx D, Monton A et al. Factors Koch AL. Orientation of the peptidoglycan chains in the sacculus
essential for L, D-transpeptidase-mediated peptidoglycan of Escherichia coli. Res Microbiol 1998;149:689–701.
cross-linking and β-lactam resistance in Escherichia coli. eLife Koch AL. Simulation of the conformation of the murein fabric:
2016;5:e19469. the oligoglycan, penta-muropeptide, and cross-linked nona-
Hugonnet J-E, Tremblay LW, Boshoff HI et al. Meropenem- muropeptide. Arch Microbiol 2000;174:429–39.
clavulanate is effective against extensively drug-resistant Korfmann G, Sanders CC. ampG is essential for high-level
Mycobacterium tuberculosis. Science 2009;323:1215–8. expression of AmpC beta-lactamase in Enterobacter cloacae.
Humann J, Lenz LL. Bacterial peptidoglycan degrading enzymes Antimicrob Agents Chemother 1989;33:1946–51.
and their impact on host muropeptide detection. J Innate Kotani S, Yanagida I, Kato K et al. Studies on peptides, glycopep-
Immun 2009;1:88–97. tides and antigenic polysaccharide-glycopeptide complexes
Hussain S, Wivagg CN, Szwedziak P et al. MreB filaments align isolated from an L-11 enzyme lysate of the cell walls of
along greatest principal membrane curvature to orient cell Mycobacterium tuberculosis strain H37Rv. Biken J 1970;13:249–
wall synthesis. eLife 2018;7:e32471. 75.
Maitra et al. 571

Koul A, Vranckx L, Dhar N et al. Delayed bactericidal response Loessner MJ, Kramer K, Ebel F et al. C-terminal domains of Lis-
of Mycobacterium tuberculosis to bedaquiline involves remod- teria monocytogenes bacteriophage murein hydrolases deter-
elling of bacterial metabolism. Nature communications mine specific recognition and high-affinity binding to bacte-
2014;5:3369. rial cell wall carbohydrates. Mol Microbiol 2002;44:335–49.
Kristan K, Kotnik M, Oblak M et al. New high-throughput Machowski EE, Senzani S, Ealand C et al. Comparative genomics
fluorimetric assay for discovering inhibitors of UDP-N- for mycobacterial peptidoglycan remodelling enzymes
acetylmuramyl-L-alanine: D-glutamate (MurD) ligase. J reveals extensive genetic multiplicity. BMC Microbiol
Biomol Screen 2009;14:412–8. 2014;14:75.
Kuk AC, Mashalidis EH, Lee SY. Crystal structure of the MOP flip- Mahajan R. Bedaquiline: first FDA-approved tuberculosis drug in
pase MurJ in an inward-facing conformation. Nat Struct Mol 40 years. Int J Appl Basic Med Res 2013;3:1–2.
Biol 2017;24:171–6. Mahapatra S, Bhakta S, Ahamed J et al. Characterization of
Kumar A, Kumar S, Kumar D et al. The structure of Rv3717 reveals derivatives of the high-molecular-mass penicillin-binding
a novel amidase from Mycobacterium tuberculosis. Acta Crystal- protein (PBP) 1 of Mycobacterium leprae. Biochem J 2000;350 Pt
logr Sec D, Biol Crystallogr 2013;69:2543–54. 1:75–80.
Kumar P, Arora K, Lloyd JR et al. Meropenem inhibits D, D- Mahapatra S, Crick DC, McNeil MR et al. Unique structural fea-
carboxypeptidase activity in Mycobacterium tuberculosis. Mol tures of the peptidoglycan of Mycobacterium leprae. J Bacteriol
Microbiol 2012;86:367–81. 2008;190:655–61.
Kumar V, Saravanan P, Arvind A et al. Identification of hotspot Mahapatra S, Scherman H, Brennan PJ et al. N Glycolylation of
regions of MurB oxidoreductase enzyme using homology the nucleotide precursors of peptidoglycan biosynthesis of
modeling, molecular dynamics and molecular docking tech- Mycobacterium spp. is altered by drug treatment. J Bacteriol
niques. J Mol Model 2011;17:939–53. 2005a;187:2341–7.
Lam H, Oh DC, Cava F et al. D-amino acids govern stationary Mahapatra S, Yagi T, Belisle JT et al. Mycobacterial lipid II is
phase cell wall remodeling in bacteria. Science 2009;325:1552– composed of a complex mixture of modified muramyl and
5. peptide moieties linked to decaprenyl phosphate. J Bacteriol
Lavollay M, Arthur M, Fourgeaud M et al. The peptidoglycan of 2005b;187:2747–57.
stationary-phase Mycobacterium tuberculosis predominantly Manjunatha U, Boshoff HI, Barry CE. The mechanism of action
contains cross-links generated by L,D-transpeptidation. J of PA-824: novel insights from transcriptional profiling. Com-
Bacteriol 2008;190:4360–6. mun Integr Biol 2009;2:215–8.
Leclercq S, Derouaux A, Olatunji S et al. Interplay between Mansour TS, Caufield CE, Rasmussen B et al. Naphthyl tetronic
penicillin-binding proteins and SEDS proteins promotes bac- acids as multi-target inhibitors of bacterial peptidoglycan
terial cell wall synthesis. Sci Rep 2017;7:43306. biosynthesis. Chem Med Chem 2007;2:1414–7.
Lee TK, Huang KC. The role of hydrolases in bacterial cell-wall Marmor S, Petersen CP, Reck F et al. Biochemical characteriza-
growth. Curr Opin Microbiol 2013;16:760–6. tion of a phosphinate inhibitor of Escherichia coli MurC. Bio-
Legaree BA, Clarke AJ. Interaction of penicillin-binding protein chemistry 2001;40:12207–14.
2 with soluble lytic transglycosylase B1 in Pseudomonas Marshall DD, Halouska S, Zinniel DK et al. Assessment of
aeruginosa. J Bacteriol 2008;190:6922–6. metabolic changes in Mycobacterium smegmatis wild-type and
Leisico F, D VV, Figueiredo TA et al. First insights of peptidoglycan alr mutant strains: evidence of a new pathway of d-alanine
amidation in Gram-positive bacteria - the high-resolution biosynthesis. J Proteome Res 2017;16:1270–9.
crystal structure of Staphylococcus aureus glutamine amido- Mauck J, Chan L, Glaser L. Turnover of the cell wall of Gram-
transferase GatD. Sci Rep 2018;8:5313. positive bacteria. J Biol Chem 1971;246:1820–7.
LeMagueres P, Im H, Ebalunode J et al. The 1.9 A crystal structure Mavrici D, Prigozhin DM, Alber T. Mycobacterium tuberculosis RpfE
of alanine racemase from Mycobacterium tuberculosis con- crystal structure reveals a positively charged catalytic cleft.
tains a conserved entryway into the active site. Biochemistry Protein Sci 2014;23:481–7.
2005;44:1471–81. Medzhitov R, Janeway CA. Innate immunity: the virtues of a non-
Levefaudes M, Patin D, De Sousa-D’Auria C et al. Diaminopimelic clonal system of recognition. Cell 1997;91:295–8.
acid amidation in Corynebacteriales: new insights into the Meeske AJ, Riley EP, Robins WP et al. SEDS proteins are a
role of LtsA in peptidoglycan modification. J Biol Chem 2015: widespread family of bacterial cell wall polymerases. Nature
jbc. M115. 642843. 2016;537:634.
Liechti G, Singh R, Rossi PL et al. Chlamydia trachomatis dapF Mengin-Lecreulx D, Falla T, Blanot D et al. Expression of
encodes a bifunctional enzyme capable of both d-glutamate the Staphylococcus aureus UDP-N-acetylmuramoyl- L-alanyl-
racemase and diaminopimelate epimerase activities. mBio D-glutamate:L-lysine ligase in Escherichia coli and effects
2018;9:e00204–18. on peptidoglycan biosynthesis and cell growth. J Bacteriol
Liger D, Masson A, Blanot D et al. Study of the overpro- 1999;181:5909–14.
duced uridine-diphosphate-N-acetylmuramate:L-alanine Meniche X, Otten R, Siegrist MS et al. Subpolar addition of new
ligase from Escherichia coli. Microb Drug Resist 1996;2: cell wall is directed by DivIVA in mycobacteria. Proc Natl Acad
25–7. Sci 2014;111:E3243–E51.
Li S, Kang J, Yu W et al. Identification of M. tuberculosis Rv3441c Meroueh SO, Bencze KZ, Hesek D et al. Three-dimensional struc-
and M. smegmatis MSMEG 1556 and essentiality of M. smeg- ture of the bacterial cell wall peptidoglycan. Proc Natl Acad Sci
matis MSMEG 1556. PLoS One 2012;7:e42769. U S A 2006;103:4404–9.
Li ZS, Beveridge TJ, Betts J et al. Partial characterization of Mikusova K, Slayden RA, Besra GS et al. Biogenesis of the
a major autolysin from Mycobacterium phlei. Microbiology mycobacterial cell wall and the site of action of ethambutol.
1999;145:169–76. Antimicrob Agents Chemother 1995;39:2484–9.
572 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

Milligan DL, Tran SL, Strych U et al. The alanine racemase Nampoothiri KM, Rubex R, Patel AK et al. Molecular cloning, over-
of Mycobacterium smegmatis is essential for growth in the expression and biochemical characterization of hypothetical
absence of D-alanine. J Bacteriol 2007;189:8381–6. beta-lactamases of Mycobacterium tuberculosis H37Rv. J Appl
Mingorance J, Tamames J, Vicente M. Genomic channeling in Microbiol 2008;105:59–67.
bacterial cell division. J Mol Recognit 2004;17:481–7. Ngadjeua F, Braud E, Saidjalolov S et al. Critical impact of
Mir M, Asong J, Li X et al. The extracytoplasmic domain of the peptidoglycan precursor amidation on the activity of L,D-
Mycobacterium tuberculosis Ser/Thr kinase PknB binds spe- Transpeptidases from Enterococcus faecium and M y cobac-
cific muropeptides and is required for PknB localization. PLoS terium tuberculosis. Chem Eur J 2018;24:5743–7.
Pathog 2011;7:e1002182. Nikitushkin VD, Demina GR, Shleeva MO et al. Peptidogly-
Mizyed S, Oddone A, Byczynski B et al. UDP-N-acetylmuramic can fragments stimulate resuscitation of “non-culturable”
acid (UDP-MurNAc) is a potent inhibitor of MurA mycobacteria. Antonie Van Leeuwenhoek 2013;103: 37–46.
(enolpyruvyl-UDP-GlcNAc synthase). Biochemistry Nikolaidis I, Izoré T, Job V et al. Calcium-dependent complex
2005;44:4011–7. formation between PBP2 and lytic transglycosylase SltB1 of
Mohammadi T, Sijbrandi R, Lutters M et al. Specificity of the Pseudomonas aeruginosa. Microb Drug Resist 2012;18:298–
transport of lipid II by FtsW in Escherichia coli. J Biol Chem 305.
2014;289:14707–18. Noldeke ER, Muckenfuss LM, Niemann V et al. Structural basis
Mohammadi T, van Dam V, Sijbrandi R et al. Identification of of cell wall peptidoglycan amidation by the GatD/MurT com-
FtsW as a transporter of lipid-linked cell wall precursors plex of Staphylococcus aureus. Sci Rep 2018;8:12953.
across the membrane. EMBO J 2011;30: 1425–32. Otten C, Brilli M, Vollmer W et al. Peptidoglycan in obligate intra-
Morayya S, Awasthy D, Yadav R et al. Revisiting the essential- cellular bacteria. Mol Microbiol 2018;107:142–63.
ity of glutamate racemase in Mycobacterium tuberculosis. Gene Pandey AK, Yang Y, Jiang Z et al. NOD2, RIP2 and IRF5 play a crit-
2015;555:269–76. ical role in the type I interferon response to Mycobacterium
Morlot C, Straume D, Peters K et al. Structure of the essential tuberculosis. PLoS Pathog 2009;5:e1000500.
peptidoglycan amidotransferase MurT/GatD complex from Parikh A, Verma SK, Khan S et al. PknB-mediated phosphoryla-
Streptococcus pneumoniae. Nature Commun 2018;9:3180. tion of a novel substrate, N-acetylglucosamine-1-phosphate
Morlot C, Uehara T, Marquis KA et al. A highly coordinated cell uridyltransferase, modulates its acetyltransferase activity. J
wall degradation machine governs spore morphogenesis in Mol Biol 2009;386:451–64.
Bacillus subtilis. Genes Dev 2010;24:411–22. Park JT, Uehara T. How bacteria consume their own exoskeletons
Mortuza R, Aung HL, Taiaroa G et al. Overexpression of a newly (turnover and recycling of cell wall peptidoglycan). Microbiol
identified d-amino acid transaminase in Mycobacterium smeg- Mol Biol Rev 2008;72: 211–27, table of contents.
matis complements glutamate racemase deletion. Mol Micro- Park JT. Uridine-5’-pyrophosphate derivatives. II. Isolation from
biol 2018;107:198–213. Staphylococcus aureus. J Biol Chem 1952;194: 877–84.
Morè N, Martorana AM, Biboy J et al. Peptidoglycan remodel- Patru MM, Pavelka MS, Jr. A role for the class A penicillin-binding
ing enables escherichia coli to survive severe outer membrane protein PonA2 in the survival of Mycobacterium smegmatis
assembly defect. mBio 2019;10:e02729–18. under conditions of nonreplication. J Bacteriol 2010;192:3043–
Moynihan PJ, Cadby IT, Veerapen Net al. The hydrolase LpqI 54.
primes mycobacterial peptidoglycan recycling. Nat Commun Payen M, Muylle I, Vandenberg O et al. Meropenem-clavulanate
2019; 10:2647. for drug-resistant tuberculosis: a follow-up of relapse-free
Mukamolova GV, Kaprelyants AS, Young DI et al. A bacterial cases. Int J Tuberc Lung Dis 2018;22:34–9.
cytokine. Proc Natl Acad Sci U S A 1998a;95:8916–21. Payne DJ, Gwynn MN, Holmes DJ et al. Drugs for bad bugs: con-
Mukamolova GV, Kormer SS, Kell DB et al. Stimulation of the fronting the challenges of antibacterial discovery. Nat Rev
multiplication of Micrococcus luteus by an autocrine growth Drug Discovery 2007;6:29–40.
factor. Arch Microbiol 1999;172:9–14. Perdih A, Hrast M, Barreteau H et al. Benzene-1,3-dicarboxylic
Mukamolova GV, Murzin AG, Salina EG et al. Muralytic activity acid 2,5-dimethylpyrrole derivatives as multiple inhibitors
of Micrococcus luteus Rpf and its relationship to physiological of bacterial Mur ligases (MurC-MurF). Bioorganic Med Chem
activity in promoting bacterial growth and resuscitation. Mol 2014;22:4124–34.
Microbiol 2006;59:84–98. Pesnot T, Gershater MC, Ward JM et al. Phosphate mediated
Mukamolova GV, Turapov OA, Kazarian K et al. The rpf gene of biomimetic synthesis of tetrahydroisoquinoline alkaloids.
Micrococcus luteus encodes an essential secreted growth fac- Chem Commun 2011;47:3242–4.
tor. Mol Microbiol 2002;46:611–21. Petit JF, Adam A, Wietzerbin-Falszpan J et al. Chemical structure
Mukamolova GV, Yanopolskaya ND, Kell DB et al. On resuscita- of the cell wall of Mycobacterium smegmatis. I. Isolation and
tion from the dormant state of Micrococcus luteus. Antonie Van partial characterization of the peptidoglycan. Biochem Biophys
Leeuwenhoek 1998b;73:237–43. Res Commun 1969;35:478–85.
Munch D, Roemer T, Lee SH et al. Identification and in Pink D, Moeller J, Quinn B et al. On the architecture of the gram-
vitro analysis of the GatD/MurT enzyme-complex catalyz- negative bacterial murein sacculus. J Bacteriol 2000;182:5925–
ing lipid II amidation in Staphylococcus aureus. PLoS Pathog 30.
2012;8:e1002509. Pisabarro AG, de Pedro MA, Vazquez D. Structural modifica-
Munshi T, Gupta A, Evangelopoulos D et al. Characterisation of tions in the peptidoglycan of Escherichia coli associated with
ATP-Dependent Mur Ligases Involved in the Biogenesis of changes in the state of growth of the culture. J Bacteriol
Cell Wall Peptidoglycan in Mycobacterium tuberculosis. PLoS 1985;161:238–42.
One 2013;8:e60143. Piuri M, Hatfull GF. A peptidoglycan hydrolase motif within
Nakel M, Ghuysen JM, Kandler O. Wall peptidoglycan in Aerococ- the mycobacteriophage TM4 tape measure protein promotes
cus viridans strains 201 Evans and ATCC 11563 and in Gaffkya efficient infection of stationary phase cells. Mol Microbiol
homari strain ATCC 10400. Biochemistry 1971;10:2170–5. 2006;62:1569–85.
Maitra et al. 573

Plocinski P, Ziolkiewicz M, Kiran M et al. Characterization Röse L, Kaufmann SH, Daugelat S. Involvement of Mycobac-
of CrgA, a new partner of the Mycobacterium tubercu- terium smegmatis undecaprenyl phosphokinase in
losis peptidoglycan polymerization complexes. J Bacteriol biofilm and smegma formation. Microbes Infect 2004;6:
2011;193:3246–56. 965–71.
Poen S, Nakatani Y, Opel-Reading HK et al. Exploring the struc- Sangshetti JN, Joshi SS, Patil RH et al. Mur Ligase Inhibitors
ture of glutamate racemase from Mycobacterium tuberculosis as Anti-bacterials: A Comprehensive Review. Curr Pharm Des
as a template for anti-mycobacterial drug discovery. Biochem 2017;23:3164–96.
J 2016;473:1267–80. Sassetti CM, Boyd DH, Rubin EJ. Genes required for mycobacte-
Pompeo F, van Heijenoort J, Mengin-Lecreulx D. Probing the rial growth defined by high density mutagenesis. Mol Micro-
role of cysteine residues in glucosamine-1-phosphate acetyl- biol 2003;48:77–84.
transferase activity of the bifunctional GlmU protein from Sauvage E, Kerff F, Terrak M et al. The penicillin-binding proteins:
Escherichia coli: site-directed mutagenesis and characteriza- structure and role in peptidoglycan biosynthesis. FEMS Micro-
tion of the mutant enzymes. J Bacteriol 1998;180:4799–803. biol Rev 2008;32:234–58.
Prigozhin DM, Mavrici D, Huizar JP et al. Structural and biochem- Scheffers DJ, Pinho MG. Bacterial cell wall synthesis: new
ical analyses of Mycobacterium tuberculosis N-acetylmuramyl- insights from localization studies. Microbiol Mol Biol Rev
L-alanine amidase Rv3717 point to a role in peptidoglycan 2005;69:585–607.
fragment recycling. J Biol Chem 2013;288:31549–55. Scheurwater E, Reid CW, Clarke AJ. Lytic transglycosylases:
Prosser GA, de Carvalho LP. Kinetic mechanism and inhibition of bacterial space-making autolysins. Int J Biochem Cell Biol
Mycobacterium tuberculosis D-alanine:D-alanine ligase by the 2008;40:586–91.
antibiotic D-cycloserine. FEBS J 2013;280:1150–66. Schubert K, Sieger B, Meyer F et al. The antituberculosis drug
Prosser GA, Rodenburg A, Khoury H et al. Glutamate racemase ethambutol selectively blocks apical growth in CMN group
is the primary target of β-chloro-D-alanine in Mycobacterium bacteria. MBio 2017;8.
tuberculosis. Antimicrob Agents Chemother 2016: AAC. 01249-16. Schulbach MC, Brennan PJ, Crick DC. Identification of a
Quesniaux V, Fremond C, Jacobs M et al. Toll-like receptor path- short (C15) chain Z-isoprenyl diphosphate synthase and
ways in the immune responses to mycobacteria. Microbes a homologous long (C50) chain isoprenyl diphosphate
Infect 2004;6:946–59. synthase in Mycobacterium tuberculosis. J Biol Chem 2000:
Radkov AD, Hsu Y-P, Booher G et al. Imaging bacterial cell wall 22876-81.
biosynthesis. Annu Rev Biochem 2018;87:991–1014. Schulbach MC, Mahapatra S, Macchia M et al. Purification,
Raymond JB, Mahapatra S, Crick DC et al. Identification of the enzymatic characterization, and inhibition of the Z-farnesyl
namH gene, encoding the hydroxylase responsible for the N- diphosphate synthase from Mycobacterium tuberculosis. J Biol
glycolylation of the mycobacterial peptidoglycan. J Biol Chem Chem 2001;276: 11624–30.
2005;280:326–33. Schulz MN, Hubbard RE. Recent progress in fragment-based lead
Reck F, Marmor S, Fisher S et al. Inhibitors of the bacterial cell discovery. Curr Opin Pharmacol 2009;9:615–21.
wall biosynthesis enzyme MurC. Bioorganic Med Chem Letters Shah IM, Laaberki MH, Popham DL et al. A eukaryotic-like
2001;11:1451–4. Ser/Thr kinase signals bacteria to exit dormancy in response
Reith J, Mayer C. Peptidoglycan turnover and recycling in Gram- to peptidoglycan fragments. Cell 2008;135:486–96.
positive bacteria. Appl Microbiol Biotechnol 2011;92:1–11. Sham L-T, Butler EK, Lebar MD et al. MurJ is the flippase of
Riano F, Arroyo L, Paris S et al. T cell responses to DosR and Rpf lipid-linked precursors for peptidoglycan biogenesis. Science
proteins in actively and latently infected individuals from 2014;345:220–2.
Colombia. Tuberculosis (Edinb) 2012;92:148–59. Shanmugam A, Anbazhagan V, Natarajan J. Virtual screen-
Rodriguez-Rivera FP, Zhou X, Theriot JA et al. Acute Modula- ing of phenylsulfonamido-3-morpholinopropan-2-yl dihy-
tion of Mycobacterial Cell Envelope Biogenesis by Front-Line drogen phosphate derivatives as novel inhibitors of MurC–
Tuberculosis Drugs. Angew Chem Int Ed 2018;57:5267–72. MurF ligases from Mycobacterium leprae. Med Chem Res
Romano M, Aryan E, Korf H et al. Potential of Mycobacterium tuber- 2012;21:4341–51.
culosis resuscitation-promoting factors as antigens in novel Shanmugam A, Natarajan J. Comparative modeling of UDP-N-
tuberculosis sub-unit vaccines. Microbes Infect 2012;14:86–95. acetylmuramoyl-glycyl-D-glutamate-2, 6-diaminopimelate
Romeis T, Holtje JV. Penicillin-binding protein 7/8 of Escherichia ligase from Mycobacterium leprae and analysis of its binding
coli is a DD-endopeptidase. Eur J Biochem 1994;224:597–604. features through molecular docking studies. J Mol Model
Roychowdhury A, Wolfert MA, Boons GJ. Synthesis and proin- 2012;18:115–25.
flammatory properties of muramyl tripeptides contain- Shetty A, Dick T. Mycobacterial cell wall synthesis inhibitors
ing lysine and diaminopimelic acid moieties. ChemBioChem cause lethal ATP burst. Front Microbiol 2018;9:1898.
2005;6:2088–97. Shrivastava P, Navratna V, Silla Y et al. Inhibition of Mycobac-
Ruggiero A, Marasco D, Squeglia F et al. Structure and func- terium tuberculosis dihydrodipicolinate synthase by alpha-
tional regulation of RipA, a mycobacterial enzyme essential ketopimelic acid and its other structural analogues. Sci Rep
for daughter cell separation. Structure 2010;18:1184–90. 2016;6:30827.
Ruiz N. Bioinformatics identification of MurJ (MviN) as the pep- Siegrist MS, Whiteside S, Jewett JC et al. D-amino acid chemical
tidoglycan lipid II flippase in Escherichia coli. Proc Natl Acad reporters reveal peptidoglycan dynamics of an intracellular
Sci 2008;105:15553–7. pathogen. ACS Chem Biol 2013;8: 500–5.
Russell-Goldman E, Xu J, Wang X et al. A Mycobacterium tubercu- Siewert G, Strominger JL. Bacitracin: an inhibitor of the dephos-
losis Rpf double-knockout strain exhibits profound defects in phorylation of lipid pyrophosphate, an intermediate in the
reactivation from chronic tuberculosis and innate immunity biosynthesis of the peptidoglycan of bacterial cell walls. Proc
phenotypes. Infect Immun 2008;76:4269–81. Natl Acad Sci 1967;57:767.
Ryan NJ, Lo JH. Delamanid: first global approval. Drugs 2014;74: Silver LL. Challenges of antibacterial discovery. Clin Microbiol Rev
1041–5. 2011;24:71–109.
574 FEMS Microbiology Reviews, 2019, Vol. 43, No. 5

Silver LL. Does the cell wall of bacteria remain a viable source of Turapov O, Forti F, Kadhim B et al. Two Faces of CwlM, an Essen-
targets for novel antibiotics? Biochem Pharmacol 2006;71:996– tial PknB Substrate, in Mycobacterium tuberculosis. Cell reports
1005. 2018;25:57–67 e5.
Singh PP, Smith VL, Karakousis PC et al. Exosomes isolated from Turk S, Kovac A, Boniface A et al. Discovery of new inhibitors of
mycobacteria-infected mice or cultured macrophages can the bacterial peptidoglycan biosynthesis enzymes MurD and
recruit and activate immune cells in vitro and in vivo. J MurF by structure-based virtual screening. Bioorganic Med
Immunol 2012;189:777–85. Chem 2009;17:1884–9.
Siricilla S, Mitachi K, Skorupinska-Tudek K et al. Biosynthesis of Turner RD, Mesnage S, Hobbs JK et al. Molecular imaging of gly-
a water-soluble lipid I analogue and a convenient assay for can chains couples cell-wall polysaccharide architecture to
translocase I. Anal Biochem 2014;461: 36–45. bacterial cell morphology. Nature Commun 2018;9:1263.
Smith CA. Structure, function and dynamics in the mur family Udou T, Ogawa M, Mizuguchi Y. Spheroplast formation of
of bacterial cell wall ligases. J Mol Biol 2006;362:640–55. Mycobacterium smegmatis and morphological aspects of their
Soni V, Upadhayay S, Suryadevara P et al. Depletion of M. tubercu- reversion to the bacillary form. J Bacteriol 1982;151:1035–9.
losis GlmU from Infected Murine Lungs Effects the Clearance Uehara A, Fujimoto Y, Kawasaki A et al. Meso-diaminopimelic
of the Pathogen. PLoS Pathog 2015;11:e1005235. acid and meso-lanthionine, amino acids specific to bacte-
Stamper GF, Longenecker KL, Fry EH et al. Structure-based rial peptidoglycans, activate human epithelial cells through
optimization of MurF inhibitors. Chem Biol Drug Design NOD1. J Immunol 2006;177:1796–804.
2006;67:58–65. Usha V, Dover LG, Roper DI et al. Structure of the diaminopime-
Strancar K, Blanot D, Gobec Set al. Design, synthesis and late epimerase DapF from Mycobacterium tuberculosis. Acta
structure-activity relationships of new phosphinate Crystallogr Sec D 2009;65:383–7.
inhibitors of MurD. Bioorg. Med. Chem. Lett. 2006; 16:343– Usha V, Lloyd AJ, Lovering AL et al. Structure and function of
8. 16271472 Mycobacterium tuberculosis meso-diaminopimelic acid (DAP)
Strancar K, Boniface A, Blanot D et al. Phosphinate inhibitors of biosynthetic enzymes. FEMS Microbiol Lett 2012;330:10–6.
UDP-N-acetylmuramoyl-L-alanyl-D-glutamate: L-lysine lig- Usha V, Lloyd AJ, Roper DI et al. Reconstruction of
ase (MurE). Arch Pharm (Weinheim) 2007;340:127–34. diaminopimelic acid biosynthesis allows characterisation of
Sulzenbacher G, Gal L, Peneff C et al. Crystal structure of Strepto- Mycobacterium tuberculosis N-succinyl-L, L-diaminopimelic
coccus pneumoniae N-acetylglucosamine-1-phosphate uridyl- acid desuccinylase. Sci Rep 2016;6:23191.
transferase bound to acetyl-coenzyme A reveals a novel Vaganay S, Tanner ME, van Heijenoort J et al. Study of the reac-
active site architecture. J Biol Chem 2001;276:11844–51. tion mechanism of the D-glutamic acid-adding enzyme from
Sutcliffe IC, Harrington DJ. Lipoproteins of Mycobacterium tuber- Escherichia coli. Microb Drug Resist 1996;2:51–4.
culosis: an abundant and functionally diverse class of cell van Asselt EJ, Thunnissen AM, Dijkstra BW. High resolution
envelope components. FEMS Microbiol Rev 2004;28:645–59. crystal structures of the Escherichia coli lytic transglycosylase
Taguchi A, Welsh MA, Marmont LS et al. FtsW is a peptidoglycan Slt70 and its complex with a peptidoglycan fragment. J Mol
polymerase that is functional only in complex with its cog- Biol 1999;291:877–98.
nate penicillin-binding protein. Nature Microbiol 2019:4:587– van Heijenoort J. Formation of the glycan chains in the synthesis
94. of bacterial peptidoglycan. Glycobiology 2001a;11:25R–36R.
Takayama K, Schnoes HK, Semmler EJ. Characterization of the van Heijenoort J. Lipid intermediates in the biosynthesis of bac-
alkali-stable mannophospholipids of Mycobacterium smegma- terial peptidoglycan. Microbiol Mol Biol Rev 2007;71:620–35.
tis. Biochem Biophys Acta 1973;316:212–21. van Heijenoort J. Recent advances in the formation of the bacte-
Talukder AA, Yanai S, Nitta T et al. RpoS-dependent regulation rial peptidoglycan monomer unit. Nat Prod Rep 2001b;18:503–
of genes expressed at late stationary phase in Escherichia coli. 19.
FEBS Lett 1996;386:177–80. Vicente M, Rico AI, Martinez-Arteaga R et al. Septum enlight-
Tanner ME, Vaganay S, van Heijenoort J et al. Phosphinate enment: assembly of bacterial division proteins. J Bacteriol
inhibitors of the D-glutamic acid-adding enzyme of peptido- 2006;188:19–27.
glycan biosynthesis. J Org Chem 1996;61:1756–60. Vijayrajratnam S, Pushkaran AC, Balakrishnan A et al. Bacte-
Templin MF, Ursinus A, Holtje JV. A defect in cell wall recy- rial peptidoglycan with amidated meso-diaminopimelic acid
cling triggers autolysis during the stationary growth phase evades NOD1 recognition: an insight on NOD1 structure-
of Escherichia coli. EMBO J 1999;18:4108–17. recognition study. Biochem J 2016:473:4573–92.
Thanky NR, Young DB, Robertson BD. Unusual features of Vincent AT, Nyongesa S, Morneau I et al. The mycobacterial cell
the cell cycle in mycobacteria: polar-restricted growth and envelope: a relict from the past or the result of recent evolu-
the snapping-model of cell division. Tuberculosis (Edinb) tion? Front Microbiol 2018;9:2341.
2007;87:231–6. Volkamer A, Eid S, Turk S et al. Pocketome of human kinases:
Tomasic T, Zidar N, Kovac A et al. 5-Benzylidenethiazolidin-4- prioritizing the ATP binding sites of (yet) untapped protein
ones as multitarget inhibitors of bacterial Mur ligases. Chem kinases for drug discovery. J Chem Inf Model 2015;55:538–49.
Med Chem 2010;5:286–95. Vollmer W, Holtje JV. The architecture of the murein (peptidogly-
Tomašić T, Mašič LP. Rhodanine as a scaffold in drug discovery: a can) in gram-negative bacteria: vertical scaffold or horizontal
critical review of its biological activities and mechanisms of layer(s)? J Bacteriol 2004;186:5978–87.
target modulation. Expert Opin Drug Discovery 2012;7:549–60. Vollmer W, Joris B, Charlier P et al. Bacterial peptidoglycan
Triboulet S, Bougault CM, Laguri C et al. Acyl acceptor recognition (murein) hydrolases. FEMS Microbiol Rev 2008;32:259–86.
by Enterococcus faecium L,D-transpeptidase Ldtfm. Mol Micro- Vollmer W, von Rechenberg M, Holtje JV. Demonstration of
biol 2015;98:90–100. molecular interactions between the murein polymerase
Trott O, Olson AJ. AutoDock Vina: improving the speed and accu- PBP1B, the lytic transglycosylase MltA, and the scaffold-
racy of docking with a new scoring function, efficient opti- ing protein MipA of Escherichia coli. J Biol Chem 1999;274:
mization, and multithreading. J Comput Chem 2010;31:455–61. 6726–34.
Maitra et al. 575

Vollmer W. Structural variation in the glycan strands of bacterial Yang Y, Severin A, Chopra Ret al. 3,5-dioxopyrazolidines, novel
peptidoglycan. FEMS Microbiol Rev 2008;32:287–306. inhibitors of UDP-N- acetylenolpyruvylglucosamine reduc-
von Rechenberg M, Ursinus A, Holtje J-V. Affinity chromatogra- tase (MurB) with activity against gram-positive bacteria.
phy as a means to study multienzyme complexes involved Antimicrob. Agents Chemother 2006;50:556–64. 16436710
in murein synthesis. Microb Drug Resist 1996;2:155–7. Yan Y, Munshi S, Leiting B et al. Crystal structure of Escherichia coli
Votyakova TV, Kaprelyants AS, Kell DB. Influence of viable cells UDPMurNAc-tripeptide d-alanyl-d-alanine-adding enzyme
on the resuscitation of dormant cells in micrococcus luteus cul- (MurF) at 2.3 A resolution. J Mol Biol 2000;304:435–45.
tures held in an extended stationary phase: the population Young DB, Perkins MD, Duncan K et al. Confronting the scien-
effect. Appl Environ Microbiol 1994;60:3284–91. tific obstacles to global control of tuberculosis. J Clin Invest
Wang Q, Matsuo Y, Pradipta AR et al. Synthesis of characteristic 2008;118:1255–65.
Mycobacterium peptidoglycan (PGN) fragments utilizing with Zapun A, Philippe J, Abrahams KA et al. In vitro reconstitution
chemoenzymatic preparation of meso-diaminopimelic acid of peptidoglycan assembly from the Gram-positive pathogen
(DAP), and their modulation of innate immune responses. Streptococcus pneumoniae. ACS Chem Biol 2013;8:2688–96.
Organic Biomol Chem 2016;14:1013–23. Zawadzke LE, Bugg TD, Walsh CT. Existence of two D-alanine:D-
Wang W, Dong C, McNeil M et al. The structural basis of chain alanine ligases in Escherichia coli: cloning and sequencing
length control in Rv1086. J Mol Biol 2008;381:129–40. of the ddlA gene and purification and characterization of the
WHO. Global Tuberculosis Report Geneva. Geneva, Switzerland: DdlA and DdlB enzymes. Biochemistry 1991;30:1673–82.
World Health Organisation, 2018. Zawadzke LE, Norcia M, Desbonnet CR et al. Identification of an
Wietzerbin J, Das BC, Petit JF et al. Occurrence of D-alanyl- inhibitor of the MurC enzyme, which catalyzes an essen-
(D)-meso-diaminopimelic acid and meso-diaminopimelyl- tial step in the peptidoglycan precursor synthesis pathway.
meso-diaminopimelic acid interpeptide linkages in the pep- Assay Drug Dev Technol 2008;6: 95–103.
tidoglycan of Mycobacteria. Biochemistry 1974;13:3471–6. Zhang Y, Heym B, Allen B et al. The catalase-peroxidase gene
Wolfert MA, Roychowdhury A, Boons G-J. Modification of the and isoniazid resistance of Mycobacterium tuberculosis. Nature
structure of peptidoglycan is a strategy to avoid detection by 1992;358:591–3.
nucleotide-binding oligomerization domain protein 1. Infect Zhang Y, Yang Y, Woods A et al. Resuscitation of dormant
Immun 2007;75:706–13. Mycobacterium tuberculosis by phospholipids or specific pep-
Workman SD, Worrall LJ, Strynadka NC. Crystal structure of an tides. Biochem Biophys Res Commun 2001;284:542–7.
intramembranal phosphatase central to bacterial cell-wall Zhang Y. The magic bullets and tuberculosis drug targets. Annu
peptidoglycan biosynthesis and lipid recycling. Nature Com- Rev Pharmacol Toxicol 2005;45:529–64.
mun 2018;9:1159. Zhang Z, Bulloch EM, Bunker RD et al. Structure and function of
Wube AA, Hüfner A, Thomaschitz C et al. Design, synthesis and GlmU from Mycobacterium tuberculosis. Acta crystallogr Sec D,
antimycobacterial activities of 1-methyl-2-alkenyl-4 (1H)- Biol Crystallogr 2009;65:275–83.
quinolones. Bioorganic Med Chem 2011;19:567–79. Zheng S, Sham LT, Rubino FA et al. Structure and mutagenic anal-
Xie J, Hodgkinson JW, Katzenback BA et al. Characterization ysis of the lipid II flippase MurJ from Escherichia coli. Proc Natl
of three Nod-like receptors and their role in antimicrobial Acad Sci U S A 2018;115:6709–14.
responses of goldfish (Carassius auratus L.) macrophages to Zidar N, Tomasic T, Sink R et al. Discovery of novel 5-
Aeromonas salmonicida and Mycobacterium marinum. Dev Comp benzylidenerhodanine and 5-benzylidenethiazolidine-2,4-
Immunol 2013;39:180–7. dione inhibitors of MurD ligase. J Med Chem 2010;53:6584–94.
Xu L, Wu D, Liu L et al. Characterization of mycobacterial UDP- Zumla A, Nahid P, Cole ST. Advances in the development of new
N-acetylglucosamine enolpyruvyle transferase (MurA). Res tuberculosis drugs and treatment regimens. Nat Rev Drug Dis-
Microbiol 2014;165:91–101. covery 2013;12:388–404.

You might also like